AU2008219972A1 - Pharmaceutical composition comprising anti-GRP78 antibody as active ingredient - Google Patents

Pharmaceutical composition comprising anti-GRP78 antibody as active ingredient Download PDF

Info

Publication number
AU2008219972A1
AU2008219972A1 AU2008219972A AU2008219972A AU2008219972A1 AU 2008219972 A1 AU2008219972 A1 AU 2008219972A1 AU 2008219972 A AU2008219972 A AU 2008219972A AU 2008219972 A AU2008219972 A AU 2008219972A AU 2008219972 A1 AU2008219972 A1 AU 2008219972A1
Authority
AU
Australia
Prior art keywords
antibody
seq
grp78
amino acid
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2008219972A
Inventor
Naoki Kimura
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Forerunner Pharma Research Co Ltd
Original Assignee
Forerunner Pharma Research Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Forerunner Pharma Research Co Ltd filed Critical Forerunner Pharma Research Co Ltd
Publication of AU2008219972A1 publication Critical patent/AU2008219972A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6843Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Description

SPECIFICATION PHARMACEUTICAL COMPOSITION COMPRISING ANTI-GRP78 ANTIBODY AS ACTIVE INGREDIENT TECHNICAL FIELD [0001] The present invention relates to a method of treating cancer treatment and anticancer agents. BACKGROUND ART [0002] GRP proteins (glucose-regulated proteins) are molecular chaperones localized to endoplasmic reticulum (ER). They are known as members of a protein family that is induced in response to various intrinsic or extrinsic ER stresses, such as glucose starvation, or accumulation of misfolded proteins in ER (Non-patent document 1). [0003] GRP78 is one of GRP proteins with a molecular weight of 78 kDa and is also well known as BiP (immunoglobulin binding protein). Overexpression, or antisense approaches directly showed that GRP78 is involved in protective role from cell death caused by ER stress (Non-patent document 1). [0004] Solid tumor cells in vivo are constantly exposed in ER stress, including glucose deprivation, hypoxia and low pH, due to a feature of the tumor microenvironment. As if to support this understanding, increased expression of GRP78 protein has been confirmed in a variety of cancer cell lines or clinical cancer specimens, correlating with malignancy (Non-patent documents 2 to 5). Further, it has been demonstrated that an overexpression of GRP78 protein is involved in the acquisition of resistance to therapeutic treatment of anticancer drugs generated by topoisomerase inhibitory activity or antiangiogenesis agents (Non-patent documents 6 and 7). In a clinical study, a group of breast cancer patients with enhanced expression of GRP78 were demonstrated to be less responsive to adriamycin-based chemotherapy than a group with lower expression of GRP78 (Non-patent document 8). [0005] These reports suggest that up-regulation of GRP78 expression in tumor is associated with the mechanisms of survival, malignant transformation, resistance to anticancer agents -1- (Non-patent document 1). [0006] GRP78 is a molecular chaperone localized in ER, whereas translocation of this molecule on the cancer cell membrane has been reported. Furthermore, the possibility of application for cancer therapy by targeting the surface-located GRP78 has been indicated by several groups, with entirely different aproach. [0007] When the rabdomisarcoma cell line TE 671/RD was treated with thapsigargin (Tg), the cell membrane was slightly stained with an anti-GRP78 antibody, as confirmed by FACS analysis, thus demonstrating cell membrane localization of the GRP78 (Non-patent document 9). [0008] It should be noted here that this report addresses a transient event during the induction of cell death by Tg treatment and that it does not show data for a persistent change in GRP78 localization in cancer cells. In addition, since the antibody used in the report is a commercially available goat-derived polyclonal antibody, its whose epitope is unknown. [0009] Later, another research group reported that the two GRP78 binding peptides (WIFPWIQL and WDLAWMFRLPVG) which acquired by phage binding assays could bind to the cell surface of the prostate cancer cell line DU145 and being internalized into the cells (Non-patent document 10). [0010] In addition, those GRP78 binding peptides fused to the cell death-inducing motif
(KLAKLAK)
2 (Non-patent document 11) have shown not only the induction of cell death on DU145 cells in vitro but also antitumor effects in an experiment on mouse transplant models (Non-patent document 10). [0011] Another research group reported that the surface-located GRP78 protein on vascular endothelial cells serves as a receptor for angiogenesis inhibitor Kringle 5 (K5) (Non-patent document 16). They further demonstrated that interaction of GRP78 with a recombinant K5 induces not only an inhibition of angiogenesis but also cell death on various cancer cell lines cultured under hypoxia (Non-patent document 16). [0012] Thus, the series of experiments described above have shown that peptides that bind to the surface-located GRP78 on cancer cells or vascular endothelial cells might be a useful -2tool as antitumor agents. However, it would be difficult to apply them in the development for clinical application since the site of the surface-located GRP78 protein recognized by these peptides is not known in the art. [0013] Aside from those findings, two entirely unrelated groups relied upon different approaches to report localization of GRP78 on the cell membrane. [0014] One group showed that the receptor of the activated forms of a2-macroglobulin (a 2 M*), which functioned as a growth factor in a prostate cancer cell line (1-LN) (Non-patent document 12), was GRP78 (Non-patent document 13). The discovery added a new finding that the GRP78 protein long considered to be an ER protein also functions as a receptor of the growth factor on the cell membrane. [0015] The other group studied that polyclonal antibody in the serum from prostate cancer patient recognize the peptide sequence "CNVSDKSC" (i.e., anti-CNVSDKSC antibody); they then identified that a target protein recognized by this antibody was the GRP78 protein (Non-patent document 14). Although the anti-CNVSDKSC antibody bound to cell surface, there were no matched or similar sequence "CNVSDKSC" within GRP78 sequence, suggesting that primary amino acid sequence "CNVSDKSC" contains a tertiary structural motief mimicking an epitope in surface-located GRP78. However, it was not identified the site of GRP78 which were recognized by this antibody. [0016] Later in time, another group performed a tertiary structural analysis of the peptide "CNVSDKSC" and identified the GRP78 primary amino acid sequence "LIGRTWNDPSVQQDIKFL" located at Leu 98 -Leu 15 Which forms the similar tertiary structure serving as a putative binding site. They then prepared a rabbit polyclonal antibody against this sequence and eventually confirmed that the antibody could stain the cell surface of cancer cells, i.e., the prostate cancer cell lines 1-LN and DU145 as well as the melanoma cell line DM413. It was also confirmed that the antibody, when added to the prostate cancer cell lines, had an ability to increase the intracellular calcium concentration, induce cell proliferation, and protect the cell from apoptosis induced by TNF-a, as observed upon addition of C 2 M* (Non-patent document 15). Since the antibody against GRP78 thusly -3mimicked the ligand activity of a 2 M*, it was revealed that the region Leu 98Leu 5 of GRP78 was an a 2 M* binding sequence (Non-patent document 15). [0017] This report validated that GRP78 is localized to cell surface in prostate cancer and it was further revealed that Leu -_Leul of GRP78 (LIGRTWNDPSVQQDIKFL) was exposed extracellularly as an a 2 M* binding sequence. [0018] Further, from another approach, it was reported that antibodies against the 98-115 region of GRP78 stained the cell surface of cancer cells, thus revealing that this regeion would be capable of serving as an extracellular epitope of GRP78. [0019] Thus it was found that the GRP78 protein is highly expressed in many cancer types, with a localization change on the cell membrane. However it has been difficult to develop a new therapeutic antibody targetted to the surface-located GRP78 on the basis of findings as below. First, the site of the GRP78 exposed on the cell surface, which were recognaized by the above-described GRP78 binding peptides is not clarified, rendering it impossible to prepare antibodies that provide similar effects to the peptides; indeed, no monoclonal antibody which functionally mimic those peptides exists. Second, the antibody which recognaizes the 98-115 region of GRP78 can bind to the surface-located GRP78 of cancer cells but, at the same time, it mimics the c 2 M* growth promoting action, so this antibody cannot be expected to display an antitumor activity. [0020] Hence, it has been considered difficult to exert antitumor activity by means of the GRP78 binding antibody. Non-patent document 1: Lee AS. Trends Biochem Sci. 2001, 26, 504-10 Non-patent document 2: Patierno, et al. 1998, Cancer Res. 47, 6220-24 Non-patent document 3: Bini et al. 1997, Electrophoresis. 18, 2832-41 Non-patent document 4: Gazit et al. 1999, Breast Cancer Res. Treat. 54, 135-46 Non-patent document 5: Fernandez et al. 2000, Breast Cancer Res. Treat. 59, 15-26 Non-patent document 6: Reddy et al, J. Bio. Chem. 2003, 278, 20915-24 Non-patent document 7: Dong et al, 2005, Cancer Res. 65, 5785-5791 Non-patent document 8: Lee et al. 2006, Cancer Res. 66, 7849-7853 -4- Non-patent document 9: Delpino et al. 1998, Molecular Membrane Biology, 15, 21-26 Non-patent document 10: Arap et al. 2004, CANCER CELL. 6, 275-284 Non-patent document 11: Javadpour et al. 1996, J. Med. Chem. 39, 3107-3113 Non-patent document 12: Asplin et al. 2000, Archives of Biochemistry and Biophysics. 383, 135-141 Non-patent document 13: Misra et al. 2002, J. Biol. Chem. 277, 42082-42087 Non-patent document 14: Mintz et al. 2003, Nat Biotech. 21, 57-63 Non-patent document 15: Gonzalez-Gronow et al. 2006, Cancer Res. 66, 11424-11431 Non-patent document 16: Davidoson et al., 2005, Cancer Res. 65, 4663-4672 DISCLOSURE OF THE INVENTION PROBLEMS TO BE SOLVED BY THE INVENTION [0021] An object of the present invention is to provide novel pharmaceutical compositions using anti-GRP78 antibodies. More particularly, the present invention aims to provide a novel method of cancer treatment using anti-GRP78 antibodies, a novel cell growth suppressing agent and an anticancer agent that contain anti-GRP78 antibodies, as well as novel anti-GRP78 antibodies. MEANS FOR SOLVING THE PROBLEMS [0022] The present inventor attempted to prepare antitumor antibodies against cancer specific surface-located GRP78. To this end, it was first necessary to identify an amino acid that would be capable of serving as an epitope for the antibody exposed on the cell surface of cancer cells. So, the present inventor purified the GRP78 protein, immunized mice with the purified GRP78 protein, and selected only anti-GRP78 antibodies that stained the cell surface of cancer cells. As a result, the inventor successfully obtained an anti-GRP78 antibody that would bind specifically to the cell surface of cancer cells. Subsequently, the inventor attempted to identify the sequence that would be recognized by the obtained antibody. The analysis revealed that the antibody specifically recognized the 40 amino acids region of the -5- 376-415 region of GRP78. It was thus revealed that the 376-415 amino acid region of GRP78 was exposed extracellularly. The inventor then confirmed that the antibody recognizing this epitope was able to be internalized into the cells rapidly. Next, based on this antibody, the inventor prepared a scFv antibody attached a toxin and analyzed an in vitro cytotoxic activity in cancer cell lines; as a result, it was revealed that the obtained toxin-labeled scFv antibody specifically killed cancer cells. Furter, the inventor analyzed the antitumor activity of the antibody in a xenograft mouse models bearing cancer cell line. The transplanted tumor volume could be significantly reduced in mice treated with the antibody. These results confirmed that the antibody could exhibit the antitumor activity not only in vitro but also in vivo. These findings revealed that the antibody against an extracellular region of GRP78 was useful as an antitumor agent. [0023] Based on these findings, the inventor of the present invention has revealed to be able to solve the aforementioned problems. [0024] Specifically, the present invention provides the embodiments described in the following (1) to (28). (1) A pharmaceutical composition containing an antibody that binds to a glucose regulated protein 78 (GRP78). (2) The composition according to (1) which is an anticancer agent. (3) The composition according to (1) or (2), wherein the antibody is a monoclonal antibody. (4) The composition according to any one of (1) to (3), wherein the antibody binds to GRP78 localized on cell surfaces. (5) The composition according to any one of (1) to (4), wherein the antibody is internalized into cells expressing GRP78. (6) The composition according to any one of (1) to (5), wherein the antibody binds to the epitope depicted in SEQ ID NO: 3. (7) The composition according to any one of (1) to (6), wherein the antibody is conjugated with a cytotoxic substance. -6- (8) A monoclonal antibody that binds to GRP78. (9) The antibody according to (8) which binds to GRP78 expressed on cell surfaces. (10) The antibody according to (8) or (9) which is internalized into cells expressing GRP78. (11) The antibody according to any one of (8) to (10) which binds to the epitope depicted in SEQ ID NO: 3. (12) The antibody according to any one of (8) to (11) which recognizes the same epitope as the one that is recognized by an antibody selected from among the following (a) to (f): (a) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 8 as CDR1, the amino acid sequence depicted in SEQ ID NO: 9 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 10 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 11 as CDR1, the amino acid sequence depicted in SEQ ID NO: 12 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 13 as CDR3; (b) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 18 as CDR1, the amino acid sequence depicted in SEQ ID NO: 19 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 20 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 21 as CDR1, the amino acid sequence depicted in SEQ ID NO: 22 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 23 as CDR3; (c) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 61 as CDR1, the amino acid sequence depicted in SEQ ID NO: 62 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 63 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 64 as CDR1, the amino acid sequence depicted in SEQ ID NO: 65 as CDR2, and the amino acid sequence depicted in -7- SEQ ID NO: 66 as CDR3; (d) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 71 as CDR1, the amino acid sequence depicted in SEQ ID NO: 72 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 73 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 74 as CDR1, the amino acid sequence depicted in SEQ ID NO: 75 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 76 as CDR3; (e) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 81 as CDR1, the amino acid sequence depicted in SEQ ID NO: 82 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 83 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 84 as CDR1, the amino acid sequence depicted in SEQ ID NO: 85 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 86 as CDR3; and (f) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 91 as CDR1, the amino acid sequence depicted in SEQ ID NO: 92 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 93 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 94 as CDR1, the amino acid sequence depicted in SEQ ID NO: 95 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 96 as CDR3. (13) The antibody according to any one of (8) to (12) which has cytotoxic activity against cells expressing GRP78. (14) The antibody according to (13) which is conjugated with a cytotoxic substance. (15) A method of delivering a cytotoxic substance into cells using an anti-GRP78 antibody. (16) A method of suppressing the growth of cells by means of a cytotoxic substance -8conjugated with an anti-GRP78 antibody. (17) The method according to (15) or (16), wherein the cells are cancer cells. (18) Use of an anti-GRP78 antibody for delivering a cytotoxic substance into cells. (19) Use of an anti-GRP78 antibody having an internalizing activity for suppressing the growth of cells. (20) The use according to (18) or (19), wherein the cells are cancer cells. (21) The use according to any one of (18) to (20), wherein the cytotoxic substance is conjugated with the anti-GRP78 antibody. (22) A process for producing a pharmaceutical composition comprising the following steps: (a) the step of providing GRP78 antibodies; (b) the step of validating whether the antibodies of (a) have an internalizing activity; (c) the step of selecting antibodies having an internalizing activity; and (d) the step of binding a cytotoxic substance to the antibodies selected in (c). (23) The process according to (22), wherein the pharmaceutical composition is an anticancer agent. (24) A method of diagnosing a cancer using an anti-GRP78 antibody. (25) The method of diagnosing according to (24) which uses an anti-GRP antibody conjugated with a labeling substance. (26) The method of diagnosis according to (24) or (25) which detects the anti-GRP78 antibody incorporated into cells.An anti-GRP78 antibody conjugated with a labeling substance. (27) A polypeptide comprising the amino acid sequence of SEQ ID NO: 3 or a fragment thereof. (28) A polypeptide comprising the amino acid sequence of SEQ ID NO: 3 or a fragment thereof. ADVANTAGES OF THE INVENTION [0025] The present invention shows that by providing novel antibodies that have an activity -9of GRP78 binding and internalization into target cells, novel pharmaceutical compositions can be provided that can be used to treat various tumors or cancers that have GRP78 exposed on the cell surface. In addition, by using antibodies having such characteristics, a method of diagnosing various tumors or cancers can be provided. BRIEF DESCRIPTION OF THE DRAWINGS [0026] FIG. 1 is a diagram showing the results of western blot analysis of the GRP78 binding activities of the obtained antibodies; a cell lysate sample prepared from DU145 cells was loaded in lane 1, and GST fused GRP78 protein purified from E. coli was loaded in lane 2, followed by staining with each antibody; AS (antiserum) is a mouse antiserum collected before cell fusion. FIG. 2 is a set of diagrams showing the results of FACS analylsis of the binding activities of the obtained anti-GRP78 antibodies to the cell surface of DU145 cell. FIG. 3 is a set of diagrams showing the results of FACS analysis of the binding activities of the GA-20 antibody to the cell surface of various cancer cells. FIG. 4(A) is a set of diagrams showing the results of FACS analysis of the binding activities of the GA-20 antibody to the cell surface of various normal or immortalized cell lines; and FIG. 4(B) is a diagram showing the results of western blot analysis of the expression of the protein GRP78 in the various normal or immortalized cell lines using GA-20. FIG. 5 is a set of diagrams showing the results of FACS analysis of the activities of the antibodies GA-20 and GA-21 for internalization into cells; each antibody was incubated with DU145 cells at 0 *C or 37 C for 2 hours, followed by detecting with a secondary antibody (FITC labeled anti-mouse IgG antibody). FIG. 6 is a set of diagrams showing the results of analysis conducted by immunocytostain to determine whether the two different anti-GRP78 antibodies (GA-20 and GA-31) would be incorporated into the cells; each antibody was added to DU145 cells under culture, and incubated for 3 hours at 37 *C; thereafter, the cells were treated in accordance with the scheme outlined below, and the antibodies incorporated into cells were analyzed. -10- FIG. 7 is a set of diagrams showing the results of western blot analysis of the epitopes of respective anti-GRP78 antibodies; the upper panel shows schematically the GST fused truncated form of GRP78 proteins (1-6) used in epitope analysis, and the lower panel shows the results of western blot. Each recombinat GST protein fused to a truncated form of GRP78 (proteins (1-6)) expressed in E. coli were subjected to SDS-PAGE followed by immunoblotting with the respective anti-GRP78 antibodies as indicated. FIG. 8 is a set of diagrams showing the results of western blotting analysis carried out to narrow down the ranges of the epitopes of GA-20 and GA-21 within GRP78; the upper panel shows schematically the GST fused to GRP78 proteins as restricted to narrower ranges, and the lower panel shows the results of western blot. The respective GST fused GRP78 proteins (1-5) are subjected to SDS-PAGE followed by immunoblotting with GA-20 and GA-21 antibodies. FIG. 9 shows the results of an analysis to detect a toxin-labeled GA-20 scFv antibody (GA20-PE40) in eluted fractions during HisTrap column purification, by an ELISA system using the GRP78 binding activity as a marker; the upper panel shows schematically the ELISA system for detecting the GRP78 binding activity of GA20-PE40, and the lower panel shows the binding activities of the eluted fractions obtained as the result of ELISA; "INITIAL" refers to the E. coli lysate induced to express GA20-PE40; "pass" refers to the fraction of the lysate that simply passed through a HisTrap column after it was applied to the column; "wash" refers to the wash fraction from the column; and "elute 1" to "elute 7" refer to the fractions eluted from the column. FIG. 10 is a set of digrams showing the cytotoxic activities of purified GA20-PE40 against DU145 cells (FIG. 10A), 22Rv1 cells (FIG. 10B), and DG44 cells (FIG. 10C); to each type of cells, eluted fractions (elutes 2, 3 or 4) were added at a concentration of 10% and, thereafter, the number of viable cells was counted and compared with the cell count in a PBS treated group to determine their percentage. FIG. 11 is a set of digrams showing the results of FACS analysis of the binding activities of the obtained anti-GRP78 antibody for the cell surface of 22Rv1 cells. - 11 - FIG. 12(A) shows schematically the GST fused GRP78 proteins used for epitope analysis on the GA-20 antibody, as well as four additional antibodies obtained by re-immunization (antibodies GC-18, GC-20, GD-4, and GD-17); FIG. 12(B) shows the retults of SDS-PAGE and CBB staining as performed to confirm that expression of the respective GST fused GRP78 proteins was induced in E. coli by IPTG supplementation. FIG. 13 is a set of diagrams showing the results of western blot analysis of the epitopes of various antibodies, in which various GST fused GRP78 proteins were subjected to SDS-PAGE for analyzing the reactivities of the respective antibodies; the table at the bottom lists the epitopes of the respective antibodies as identified from the results of western blotting analysis. FIG. 14 is a diagram showing the result of SDS-PAGE followed by CBB staining conducted on purified GD17scFv-PE40 to determine its purity. FIG. 15 shows the results of ELISA conducted to analyze both the GRP78 protein binding activities of the purified GD17scFv-PE40 and the stability of the protein; GD17scFv-PE40 stored at 4 C, or let to stand overnight at 37 C, or subjected to a freeze-thaw cycle was diluted to various concentrations and its activity for binding to GST-GRP78 was analyzed by ELISA; the table at the bottom shows the GRP78 protein binding activities of the respective specimens as EC 5 0 values. FIG. 16A shows the results of evaluating the cytotoxic activities of the purified GD17scFv-PE40 on various cell lines; GD17scFv-PE40 was diluted to various concentrations and added to cancer cell lines (FIG. 16A) which were cultured for several days, and the number of viable cells was counted; antibody concentrations
(EC
50 ) that conferred an activity equivalent to 50% of a maximum activity are listed in the table at the bottom. FIG. 16B shows the results of evaluating the cytotoxic activities of the purified GD17scFv-PE40 on various cell lines; GD17scFv-PE40 was diluted to various concentrations and added to normal cell lines (FIG. 16B) which were cultured for several days, and the number of viable cells was counted; antibody concentrations
(EC
5 o) that - 12 conferred an activity equivalent to 50% of a maximum activity (EC 5 o) are listed in the table at the bottom. FIG. 17 is a diagram showing the results of analyzing the expression of the GRP78 protein in various cell lines by western blot analysis using the GD-17 antibody. FIG. 18 shows the results of analyzing the antitumor activity of GD17scFv-PE40 in an in vivo mouse xenograft model; immediately after grafting of 22Rv1 (at day 0), or at days 17, 21, 23, 26 and 29, PBS (vehicle) or 0.5 mg/kg of GD17scFv-PE40 was administered (as indicated by the arrows) and, thereafter, the tumor volume was measured over time. BEST MODE FOR CARRYING OUT THE INVENTION [0027] The anti-GRP78 antibody of the present invention may be of any type that binds to the GRP78 protein (SEQ ID NO: 2) and it is by no means limited in such aspects as the origin (mouse, rat, human, etc.), type (monoclonal antibody or polyclonal antibody), and shape (altered antibody, small molecular antibody, modified antibody, etc.) [0028] The anti-GRP78 antibody to be used in the present invention preferably specifically binds to GRP78. It is also preferred that the anti-GRP78 antibody to be used in the present invention is a monoclonal antibody. [0029] GRP78 is known to be localized on the plasma membrane of cancer cells and the like. One preferred embodiment of the anti-GRP78 antibody to be used in the present invention may be an antibody that recognizes the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane. [0030] Such antibody can be acquired by, for example, preparing antibodies using the GRP78 protein (SEQ ID NO: 2) as an immunogen and selecting from among the prepared antibodies those which can bind to cancer cells expressing GRP78 on the cell membrane (e.g. prostate cancer cell line DU145). More specifically, the method described in the Examples may typically be employed to acquire an antibody that recognizes the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane. [0031] In the present invention, the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane is preferably such that when the antibody binds to that - 13 region, it will not mimic the growth promoting action of a 2 macroglobulin and a particularly preferred region is other than the 9 8 th to 1 15 th region of GRP78. [0032] Preferred examples of the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane include that region of the amino acid sequence depicted by SEQ ID NO: 2 which is between the 3 7 6 th and the 4 15 th position (SEQ ID NO: 3). Therefore, preferred examples of an antibody that recognizes the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane include those antibodies that recognize the 3 7 6 th to 4 15 th region of GRP78. Examples of the antibody that recognizes the 376 to 415 amino acids in the protein GRP78 are not particularly limited and include an antibody that recognizes the 3 84 th to 391" amino acids (i.e., amino acids 9-16 in SEQ ID NO: 3), an antibody that recognizes the 3 9 2 nd to 4 0 7 th amino acids (i.e., amino acids 17-32 in SEQ ID NO: 3), and an antibody that recognizes the 4 0 0 th to 4 1 5 th amino acids (i.e., amino acids 25-40 in SEQ ID NO: 3). Whether a particular antibody recognizes the intended epitope can be confirmed by methods known to skilled artisans, for example, by the method described in the Examples. [0033] Other preferred embodiments of the antibody to be used in the present invention include antibodies having an internalizing activity. In the present invention, the "antibodies having an internalizing activity" means those antibodies which will be transported into cells (e.g., into the cytoplasm, vesicles or other organelles) after binding to GRP78 localized on the cell surface. [0034] Whether a particular antibody has an internalizing activity can be confirmed by methods known to skilled artisans, for example, a method in which an anti-GRP78 antibody conjugated with a labeling substance is brought into contact with cells expressing GRP78 (e.g. prostate cancer cell line DU145) and a check is made to see if the labeling substance has been incorporated into the cells, and a method in which an anti-GRP78 antibody conjugated with a cytotoxic substance is brought into contact with cells expressing GRP78 and a check is made to see if cell death has been induced in those GRP78 expressing cells. More specifically, the method described in the Examples may typically be employed to check to see if a particular - 14 antibody has an internalizing activity. [0035] In the present invention, particularly preferred antibodies include one that recognizes the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane and which has an internalizing activity. Such antibodies can be acquired by first employing the above-exemplified method to select those antibodies which recognize the region of GRP78 that is exposed extracellularly when it is localized on the cell membrane and subsequently further selecting from the thus selected antibodies those which have an internalizing activity. [0036] Examples of the preferred antibodies that may be used in the present invention include the following antibodies (a) to (s). (a) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 8 as CDR1, the amino acid sequence of SEQ ID NO :9 as CDR2, and the amino acid sequence of SEQ ID NO: 10 as CDR3. (b) An antibody comprising a light-chain variable region having the amino acid sequence of SEQ ID NO: 11 as CDR1, the amino acid sequence of SEQ ID NO: 12 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3; (c) An antibody comprising the heavy-chain variable region of (a) and the light-chain variable region of (b). (d) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 18 as CDR1, the amino acid sequence of SEQ ID NO: 19 as CDR2, and the amino acid sequence of SEQ ID NO: 20 as CDR3. (e) An antibody comprising a light-chain variable region having the amino acid sequence of SEQ ID NO: 21 as CDR1, the amino acid sequence of SEQ ID NO: 22 as CDR2, and the amino acid sequence of SEQ ID NO: 23 as CDR3. (f) An antibody comprising the heavy-chain variable region of (d) and the light-chain variable region of (e). (g) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 61 as CDR1, the amino acid sequence of SEQ ID NO: 62 as CDR2, - 15 and the amino acid sequence of SEQ ID NO: 63 as CDR3. (h) An antibody containing a light-chain variable region comprising the amino acid sequence of SEQ ID NO: 64 as CDR1, the amino acid sequence of SEQ ID NO: 65 as CDR2, and the amino acid sequence of SEQ ID NO: 66 as CDR3. (i) An antibody comprising the heavy-chain variable region of (g) and the light-chain variable region of (h). (j) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 71 as CDR1, the amino acid sequence of SEQ ID NO: 72 as CDR2, and the amino acid sequence of SEQ ID NO: 73 as CDR3. (k) An antibody comprising a light-chain variable region having the amino acid sequence of SEQ ID NO: 74 as CDR1, the amino acid sequence of SEQ ID NO: 75 as CDR2, and the amino acid sequence of SEQ ID NO: 76 as CDR3. (1) An antibody comprising the heavy-chain variable region of (j) and the light-chain variable region of (k). (m) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 81 as CDR1, the amino acid sequence of SEQ ID NO: 82 as CDR2, and the amino acid sequence of SEQ ID NO: 83 as CDR3. (n) An antibody comprising a light-chain variable region having the amino acid sequence of SEQ ID NO: 84 as CDR1, the amino acid sequence of SEQ ID NO: 85 as CDR2, and the amino acid sequence of SEQ ID NO: 86 as CDR3. (o) An antibody comprsing the heavy-chain variable region of (m) and the light-chain variable region of (n). (p) An antibody comprising a heavy-chain variable region having the amino acid sequence of SEQ ID NO: 91 as CDR1, the amino acid sequence of SEQ ID NO: 92 as CDR2, and the amino acid sequence of SEQ ID NO: 93 as CDR3. (q) An antibody comprising a light-chain variable regionhaving the amino acid sequence of SEQ ID NO: 94 as CDR1, the amino acid sequence of SEQ ID NO: 95 as CDR2, and the amino acid sequence of SEQ ID NO: 96 as CDR3. - 16 - (r) An antibody comprising the heavy-chain variable region of (p) and the light-chain variable region of (q). (s) An antibody recognizing the same epitope as what is recognized by either one of the antibodies (a) to (r). [0037] An antibody that recognizes the same epitope as a particular antibody may be obtained by the following procedure. [0038] A subject antibody can be confirmed to share the same epitope with a particular antibody by examining the competition between the two antibodies for the same epitope. Competition between the two antibodies is detected by cross-blocking assay or the like. For example, competive ELISA assay is a preferred example of the cross-blocking assay. Specifically, in cross-blocking assay, the GRP78 protein coated on the surface of wells in a microtiter plate is preincubated in the presence or absence of a candidate competing antibody and then the anti-GRP78 antibody of the present invention is added. The amount of the anti-GRP78 antibody of the present invention that binds to the GRP78 protein within the wells correlates indirectly to the binding ability of the candidate competing antibody (subject antibody) that competes for binding to the same epitope. In other words, the greater the affinity of the subject antibody for the same epitope, the smaller the amount of the anti-GRP78 antibody of the present invention that binds to the wells coated with the GRP78 protein and the greater the amount of the subject antibody that binds to the wells coated with the GRP78 protein. [0039] The amount of the antibody that has bound to the wells can be easily measured by preliminary labeling of the antibody. For example, biotin-labeled antibodies can be measured by using an avidin-peroxidase conjugate and a suitable substrate. A cross-blocking assay that utilizes an enzyme label such as peroxidase is especially referred to competitive ELISA assay. Antibodies can be labeled with other labeling substances that are detectable or measurable. Specifically, a radioactive label or a fluorescent label and the like are known. [0040] Furthermore, if the subject antibody has a constant region derived from a different species than the anti-GRP78 antibody of the present invention, the amount of the antibody -17that has bound to the wells can also be measured by a labeled antibody that recognizes the constant region of that antibody. Alternatively, the subject antibody may be derived from the same species as the anti-GRP78 antibody of the present invention but belongs to a different class; in this case, the amount of the antibody that has bound to the wells can be measured by an antibody that distinguishes between respective classes. [0041] If, compared to the binding activity obtained in a control test conducted in its absence, the candidate competing antibody can block the binding of the anti-GRP78 antibody by at least 20%, preferably at least 20-50%, more preferably at least 50%, that candidate competing antibody is either an antibody that binds to substantially the same epitope as the anti-GRP78 antibody of the present invention or an antibody that competes for the binding to the same epitope. [0042] An antibody that is conjugated with a cytotoxic substance may be mentioned as another preferred embodiment of the antibody to be used in the present invention. When the antibody that is conjugated with a cytotoxic substance is incorporated into cells, the cytotoxic substance is capable of inducing a killing action or cell death in the cells that have incorporated that antibody. Therefore, it is preferred that the antibody conjugated with a cytotoxic substance further has an internalizing activity. [0043] Preferred embodiments of the anti-GRP78 antibody that is conjugated with a cytotoxic substance according to the present invention include, for example, antibodies that have a cytotoxic activity or induce cell death on GRP78 expressing cancer cells (e.g. DU145, 22Rv1, and MCF7). [0044] The cytotoxic substance to be used in the present invention may be any substance that can induce a killing action or cell death in cells and it may be exemplified by toxins, radioactive substances, chemotherapeutics and the like. These cytotoxic substances to be used in the present invention include prodrugs that will be transformed to active cytotoxic substances in the living body. Activation of prodrugs may be generated through enzymatic or non-enzymatic transformation. [0045] The term "toxins" as used herein means various proteins, polypeptides and the like - 18 that show cytotoxicity as derived from microorganisms, animals or plants. Toxins to be used in the preent invention may include the following: Diphtheria toxin A chain (Langone J.J., et al., Methods in Enzymology, 93, 307-308, 1983); Pseudomonas exotoxin (Nature Medicine, 2, 350-353, 1996); Ricin A chain (Fulton R.J., et al., J. Biol. Chem., 261, 5314-5319, 1986; Sivam G., et al., Cancer Res., 47, 3169-3173, 1987; Cumber A.J. et al., J. Immunol. Methods, 135, 15-24, 1990; Wawrzynczak E.J., et al., Cancer Res., 50, 7519-7562, 1990; Gheeite V., et al., J. Immunol. Methods, 142, 223-230, 1991); Deglicosylated ricin A chain (Thorpe P.E., et al., Cancer Res., 47, 5924-5931, 1987); Abrin A chain (Wawrzynczak E.J., et al., Br. J. Cancer, 66, 361-366, 1992; Wawrzynczak E.J., et al., Cancer Res., 50, 7519-7562, 1990; Sivam G., et al., Cancer Res., 47, 3169-3173, 1987; Thorpe P.E., et al., Cancer Res., 47, 5924-5931, 1987); Gelonin (Sivam G., et al., Cancer Res., 47, 3169-3173, 1987; Cumber A.J. et al., J. Immunol. Methods, 135, 15-24, 1990; Wawrzynczak E.J., et al., Cancer Res., 50, 7519-7562, 1990; Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); PAP-s or pokeweed anti-viral protein from seeds (Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Briodin (Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Saporin (Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Momordin (Cumber A.J., et al., J. Immunol. Methods, 135, 15-24, 1990; Wawrzynczak E.J., et al., Cancer Res., 50, 7519-7562, 1990; Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Momorcochin (Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Dianthin 32 (Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992); Dianthin 30 (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Modeccin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Viscumin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Volkesin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Dodecandrin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Tritin (Stirpe F., Barbieri L., FEBS letter 195, 1-8, 1986); Luffin (Stirpe F., Barbieri L., FEBS letter 95, 1-8, 1986); Trichokirin (Casellas P., et al., Eur. J. Biochem. 176, 581-588, 1988; Bolognesi A., et al., Clin. Exp. Immunol., 89, 341-346, 1992). [0046] The term "radioactive substances" as used herein refers to those substances that contain radioisotopes. Radioisotopes are not particularly limited and any radioisotopes may - 19 be used; examples that can be used include "P, "C, 1I, 3H, 1, 186Re, 188Re, etc. [0047] The term "chemotherapeutics" as used herein means those substances other than the above-mentioned toxins and radioactive substances which have cytotoxic activity and it encompasses cytokines, antitumor agents, enzymes, etc. The chemotherapeutics that can be used in the present invention are not particularly limited and those of lower molecular weights are preferred. With lower molecular weights, the chemotherapeutics would be less likely to interfere with the function of the antibody even after they bind to it. In the present invention, the chemotherapeutics of lower molecular weights generally have molecular weights of 100-2000, preferably 200-1000. Although not particularly Imited, the chemotherapeutics that can be used in the present invention include the following: Melphalan (Rowland G.F., et al., Nature 255, 487-488, 1975); Cis-platinum (Hurwitz E. and Haimovich J., Methods In Enzymology 178, 369-375, 1986; Schechter B., et al., Int. J. Cancer 48, 167-172, 1991); Carboplatin (Ota, Y., et al., Asia-Oceania J. Obstet. Gynaecol. 19, 449-457, 1993); Mitomycin C (Noguchi, A., et al., Bioconjugate Chem. 3, 132-137, 1992); Adriamycin (Doxorubicin) (Shih, L.B., et al., Cancer Res. 51 4192-4198, 1991; Zhu, Z., et al., Cancer Immunol. Immumother 40, 257-267, 1995; Trail, P.A., et al., Science 261, 212-215, 1993; Kondo, Y., et al., Jpn. J. Cancer Res. 86, 1072-1079, 1995;); Daunorubicin (Dillman, R.O., et al., Cancer Res. 48, 6097-6102, 1988; Hudecz, F., et al., Bioconjugate Chem. 1, 197-204, 1990; Tukada Y. et al., J. Natl. Cancer Inst. 75, 721-729, 1984); Bleomycin (Manabe, Y., et al., Biochem. Biophys. Res. Commun. 115, 1009-1014, 1983); Neocarzinostatin (Kitamura K., et al., Cancer Immunol. Immumother 36, 177-184, 1993; Yamaguchi T., et al., Jpn. J. Cancer Res. 85, 167-171, 1994); Methotrexate (Kralovec, J., et al., Cancer Immunol. Immumother 29, 293-302, 1989; Kulkarni, P.N., et al., Cancer Res. 41, 2700-2706, 1981; Shin, L.B., et al., Int. J. Cancer 41, 832-839, 1988; Gamett M.C., et al., Int. J. Cancer 31, 661-670, 1983); 5-Fluorouridine (Shin, L.B., Int. J. Cancer 46, 1101-1106, 1990); 5-Fluoro-2'-deoxyuridine (Goerlach A., et al., Bioconjugate Chem. 2, 96-101, 1991); Cytosine arabinoside (Hurwitz E., et al., J. Med. Chem. 28, 137-140, 1985); Aminopterin (Kanellos J., et al., Immunol. Cell. Biol. 65, 483-493, 1987); vincristine (Johnson J.R., et al., - 20 - Br. J. Cancer 42, 17, 1980); vindesine (Johnson J.R., et al., Br. J. Cancer 44, 472-475, 1981); interleukine-2 (IL-2), tumor necrosis factor a (TNFa), interferon (INF), carboxypeptidase, alkaline phosphatase, P-lactamase, and cytidine deaminase. [0048] The cytotoxic substances may be used in the present invention either individually or in combination of two or more species. [0049] The anti-GRP78 antibody can be bound to the above-listed cytotoxic substances by covalent bonding, non-covalent bonding or the like. Methods of preparing antibodies conjugated with those cytotoxic substances are known. [0050] The anti-GRP78 antibody and the cytotoxic substance can be directly bound together via the linking groups they have by themselves; alternatively, they may be bound together indirectly via other substances such as linkers or intermediate supports. Examples of the linking groups that occur in the case where the anti-GRP78 antibody and the cytotoxic substance are directly bound together include a disulfide bond using the SH groups. Specifically, the intramolecular disulfide bond in the Fc region of the antibody is reduced with a reducing agent, such as dithiothreitol, and the disulfide bond within the cytotoxic substance is likewise reduced so that the two are bound by the disulfide bond. Prior to binding, either the antibody or the cytotoxic substance may be activated by an activation promoting agent, such as an Ellman's reagent so that the formation of the disulfide bond between the two molecules is accelerated. Other approaches for directly binding the anti-GRP78 antibody and the cytotoxic substance include a method using a Schiff base, a carbodiimide method, an active ester method (N-hydroxysucccinimide method), a method using a mixed anhydride, and a method using a diazo reaction. [0051] The anti-GRP78 antibody and the cytotoxic substance can also be bound together indirectly via other substances. Other substances for realizing indirect binding are not particularly limited and include, for example, compounds that have at least two substituents consisting of any one of an amino group, a carboxyl group, and a mercapto group or any combination of two or more groups, peptide linkers, and compounds having a capability of binding to the anti-GRP78 antibody. Examples of the compounds that have at least two - 21 substituents consisting of any one of an amino group, a carboxyl group, and a mercapto group or any combination of two or more groups include: N-succinimidyl 3-(2 pyridyldithio)propionate (SPDP) (Wawrzynczak E.J., et al., Cancer Res., 50, 7519-7562, 1990; Thorpe P.E., et al., Cancer Res., 47, 5924-5931, 1987); succinimidyl 6-3-[2 pyridyldithio]propionamide)hexanoate (LC-SPDP) (Hermanson G.T., BIOCONJUGATE Techniques, 230-232, 1996); sulfosuccinimidyl 6
-
3
-[
2 -pyridyldithio]propionamide)hexanoate (sulfo-LC-SPDP) (Hermanson G.T., BIOCONJUGATE Techniques, 230-232, 1996); N-Succinimidyl 3-(2-pyridyldithio)butyrate (SPDB) (Wawrzynczak E.J., et al., Br. J. Cancer, 66, 361-366, 1992); succinimidyloxycarbonyl-a-(2-pyridyldithio)toluene (SMPT) (Thorpe P.E., et al., Cancer Res., 47, 5924-5931, 1987); succinimidyl 6-(a-methyl-[2 pyridyldithio]toluamide)hexanoate (LC-SMPT) (Hermanson G.T., BIOCONJUGATE Techniques, 232-235, 1996); sulfosuccinimidyl 6-(a-methyl-[2 pyridyldithio]toluamide)hexanoate (sulfo-LC-SMPT) (Hermanson G.T., BIOCONJUGATE Techniques, 232-235, 1996); succinimidyl-4-(p-maleimidophenyl)butyrate (SMPB) (Hermanson G.T., BIOCONJUGATE Techniques, 242-243, 1996); sulfo-succinimidyl-4-(p maleimidophenyl)butyrate (sulfo-SMPB) (Hermanson G.T., BIOCONJUGATE Techniques, 242-243, 1996); m-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) (Hermanson G.T., BIOCONJUGATE Techniques, 237-238, 1996); m-maleimidobenzoyl-N hydroxysulfosuccinimide ester (sulfo-MBS) (Hermanson G.T., BIOCONJUGATE Techniques, 237-238, 1996); S-acetyl mercaptosuccinic anhydride (SAMSA) (Casellas P., et al., Eur. J. Biochem, 176, 581-588, 1988); dimethyl 3, 3'-dithiobisprorionimidate (DTBP) (Casellas P., et al., Eur. J. Biochem, 176, 581-588, 1988); and 2-iminothiolane (Thorpe P.E., et al., Cancer Res., 47, 5924-5931, 1987). [0052] Other substances that may be used to bind the anti-GRP78 antibody and the cytotoxic substance may be exemplified by peptides, antibodies, poly(L-glutamic acid) (PGA), carboxymethyldextran, dextran, aminodextran, avidin-biotin, cis-aconitic acid, glutamic acid dihydrazide, and human serum albumin (HAS). [0053] Further, proteinaceous cytotoxic substances can also be bound to the antibody by - 22 genetic engineering techniques. Specifically, a DNA coding for the above-mentioned cytotoxic peptide and a DNA coding for the anti-GRP78 antibody may be fused in frame and incorporated into an expression vector to construct a recombinant vector. This vector is then introduced into suitable host cells to generate transformed cells, which are cultured to express the incorporated DNA, whereby a fused protein of the anti-GRP78 antibody and the toxic peptide can be obtained. In the case of producing fusion proteins of the antibody and a cytotoxic substance, proteinaceous chemicals or toxins are typically positioned at the C terminus of the antibody. It is also possible to interpose a peptide linker between the antibody and the proteinaceous chemical or toxin. [0054] The anti-GRP78 monoclonal antibody of the present invention can be acquired by known means. Monoclonal antibodies derived from mammalian animals are particularly preferred as the anti-GRP78 monoclonal antibody of the present invention. The monoclonal antibodies derived from mammalian animals include, for example, those which are produced by hybridomas, as well as those which are produced from host cells that have been transformed with expression vector harboring an antibody gene by genetic engineering techniques. [0055] Monoclonal antibody producing hybridomas can typically be constructed as follows using known techniques. First, the GRP78 protein is used as a sensitizing antigen to effect immunization in accordance with a conventional immunization method. Immune cells obtained from the immunized animal are fused to known parent cells by a conventional cell fusion method to yield hybridomas. From the thus produced hybridomas, cells that produce the desired antibody are screened by a conventional screening method so as to select hybridomas that produce the anti-GRP78 protein. [0056] Specifically, the monoclonal antibody preparation can typically be performed as described below. First, the GRP78 protein (SEQ ID NO: 2) to be used as a sensitizing antigen for antibody acquisition can be acquired by expressing the GRP78 gene. The nucleotide sequence of the human GRP78 gene has been already known (SEQ ID NO: 1). To be more specific, a gene sequence coding for GRP78 is inserted into a known expression - 23 vector to transform suitable host cells and the intended human GRP78 proein can be purified from the transformed host cells or the culture supernatant by a known method. A purified native GRP78 protein can also be used. Purification can be performed by a plurality of conventional chromatographic techniques such as ion chromatography and affinity chromatography, which are used once or more than once, either individually or in combination. In addition, as in the case of the present invention, a fusion protein of a desired partial polypeptide of the GRP78 protein with a different polypeptide may be used as an immunogen. To produce the fused protein that serves as an immuogen, Fc fragments of an antibody, peptide tags and the like may be used. To construct a vector that expresses the fused protein, genes that code for two or more desired polypeptide fragments may be fused in frame and the fused genes inserted into an expression vector as described above. The method of preparing fusion proteins is described in Sambrook, J. et al., Molecular Cloning 2 "d ed., 9.47-9.58, Cold Spring Harbor Lab. Press, 1989. [0057] The thus purified GRP78 protein can be used as a sensitizing antigen to immunize mammals. Partial peptides of GRP78 can also be used as an sensitizing antigen. For example, the following peptidies can serve as sensitizing antigens: A peptide acquired by chemical synthesis from the amino acid sequence of human GRP78; A peptide acquired by incorporating part of the human GRP78 gene into an expression vector and expressing the same; A peptide acquired by decomposing the human GRP78 protein with a proteolytic enzyme. [0058] The region and size of the GRP78 to be used as the partial peptide are by no means limited. A preferred region can be selected from the 3 7 6 th to 4 15 th region of GRP78 (SEQ ID NO: 3). The peptide that serves as a sensitizing antigen is preferably composed of at least three, for exampe five or six, amino acid residues. More specifically, a peptide of 8-50 residues, preferably 10-30 residues, can be used as a sensitizing antigen. [0059] The mammals to be immunized with the sensitizing antigen are not particularly - 24 limited. To obtain a monoclonal antibody by the cell fusion method, the animal to be immunized is preferably selected in consideration of compatibility of the immunized cell with the parent cell to be used in cell fusion. In general, rodents are preferred animals to be immunized. Specifically, mouse, rat, hamster or rabbit may be used as an animal to be immunized. Other animals that may be immunized include monkey and the like. [0060] The animals mentioned above can be immunized with the sensitizing antigen in accordance with known methods. An exemplary general method comprises immunizing a mammal by intraperitoneal or subcutaneous injection of the sensitizing antigen. Specifically, the sensitizing antigen is administered to the mammal several times every 4 to 21 days. The sensitizing antigen is used for immunization after it is diluted to a suitable dilution ratio with PBS (phosphate-buffered saline), physiological saline or the like. If desired, the sensitizing antigen may be administered together with an adjuvant. For example, the sensitizing antigen may be mixed with a Freund's complete adjuvant, which may be emulsified to make a desired sensitizing antigen. In addition, a suitable carrier may be used in immunization with the sensitizing antigen. Particularly in the case where a partial peptide having a small molecular weight is used as the sensitizing antigen, immunization is preferably done by binding the sensitizing antigen peptide to a carrier protein such as albumin or keyhole limpet hemocyanine. [0061] With the mammal being thus immunized and after a increase in the amount of desired serum antibody is confirmed, the immune cells are collected from the mammal and subjected to cell fusion. Spleen cells can especially be used as preferred immune cells. [0062] The cells to be fused with the immune cells are mammalian myeloma cells. Myeloma cells are preferably furnished with a selection marker suitable for screening. The selection marker refers to a phenotype that can survive (or cannot survive) under particular culture conditions. Known selection markers include hypoxanthine-guanine-phosphoribosyl transferase deficiency (hereinafter abbreviated as HGPRT deficiency) and thymidine kinase deficiency (hereinafter abbreviated as TK deficiency). Cells having HGPRT or TK deficiency have hypoxanthine-aminopterin-thymidine sensitivity (hereinafter abbreviated as - 25 - HAT sensitivity). In a HAT selective medium, cells having HAT sensitivity are incapable of DNA synthesis and will die; however, if they are fused with normal cells, they can continue the synthesis of DNA by making use of the salvage circuit in the normal cell and hence will proliferate in the HAT selective medium. [0063] Cells with HGPRT deficiency or TK deficiency can be selected in a medium containing 6-thioguanine, 8 -azaguanine (hereinafter abbreviated as 8-AG) or 5'-bromodeoxyuridine. Normal cells incorporate these pyrimidine analogs into their DNA and will eventually die; on the other hand, cells lacking these enzymes are unable to incorporate those pyrimidine analogs and can hence survive in the selective medium. Another selection marker called G418 resistance confers resistance to 2 -deoxystreptamine antibiotics (gentamycin analogs) by means of the neomycin resistance gene. A variety of myeloma cells suitable for cell fusion are known. Myeloma cells that can be used include: P3 (P3x63Ag8.653) (J. Immunol. (1979) 123, 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (Kohler. G. and Milstein, C. Eur. J. Immunol. (1976) 6, 511-519), MPC-11 (Margulies. D.H. et al., Cell (1976) 8, 405-415), SP2/0 (Shulman, M. et al., Nature (1978) 276, 269-270), FO (de St. Groth, S. F. Et al., J. Immunol. Methods (1980) 35, 1-21), S194 (Trowbridge, I. S. J. Exp. Med. (1978) 148, 313-323), and R210 (Galfre, G. et al., Nature (1979) 277, 131-133). [0064] Cell fusion of the above-mentioned immune cells and myeloma cells can be performed in accordance with known methods, such as the method of Kohler and Milstein (Kohler. G. and Milstein, C., Methods Enzymol. (1981) 73, 3-46). [0065] More specifically, the above-mentioned cell fusion can typically be implemented in a conventional nutrient culture medium in the presence of a cell fusion promoting agent. Examples of the cell fusion promoting agent that can be used include polyethylene glycol (PEG) and Sendai virus (HVJ). If desired, an auxiliary agent such as dimethyl sulfoxide may be added in order to enhance fusion efficiency. [0066] The proportions of the immune cells and myeloma cells to be used can be set at desired values. For example, the immune cells are preferably used in amounts ranging from - 26 one to ten times the amount of myeloma cells. Culture media that can be used in the aforementioned cell fusion include, for example, the RPMI1640 culture medium and MEM culture medium suitable for the growth of the above-mentioned myeloma cell line, as well as conventional culture media used in cell culture of the type under consideration. Further, a serum supplement such as fetal calf serum (FCS) may be added to the culture medium. [0067] In cell fusion, specified amounts of the above-mentioned immune cells and myeloma cells are mixed well in the above-mentioned culture medium and then mixed with a pre-warmed (ca. 37 *C) PEG solution to form the desired fused cells (hybridomas). In the cell fusion method, PEG with an average molecular weight of from about 1000 to about 6000 can be added at concentrations typically ranging from 30 to 60% (w/v). Subsequently, procedures of seququentially adding suitable culture media as selected from those mentioned above, centrifuging them and removing the supernatant are repeated to thereby remove the cell fusion promoting agents and the like that are not preferred for the growth of hybridomas. [0068] The hybridomas thus obtained can be selected by employing a selective culture medium in accordance with the selection marker possessed by the myeloma used in cell fusion. For instance, cells having HGPRT or TK deficiency can be selected by culturing them in a HAT culture medium (i.e., containing hypoxanthine, aminopterin, and thymidine). To be more specific, in the case of using HAT sensitive myeloma cells in cell fusion, the cells that successfully fused to normal cells can be selectively grown in the HAT culture medium. The fused cells are continuously cultured using this HAT culture medium for a suifficient time that cells (nonfused cells) other than the desired hybridomas will die. Specifically, the desired hybridomas can bs selected by culturing for a period which typically ranges from several days to several weeks. Subsequently, a conventional method of limiting dilution is implemented to thereby enable the screening and a single cell cloning of hybridomas that produce the desired antibody. Alternatively, antibodies that recognize GRP78 can be constructed by the method described in WO 03/104453. [0069] Screening and cloning of the desired antibodies can advantageously be implemented by screening methods based on known antigen-antibody reactions. For instance, the antigen - 27 is bound to a carrier such as beads made of polystyrene or otherwise or a commercial 96-well microtiter plate and reacted with the culture supernatant of hybridomas. Subsequently, the carrier is washed and thereafter reacted with enzyme-labeled secondary antibodies or the like. If the culture supernatant contains the desired antibodies that react with the sensitizing antigen, the secondary antibodies indirectly bind to the carrier via the desired antibodies. Finally, the secondary antibodies indirectly binding to the carrier are detected to thereby determine whether the desired antibodies are present in the culture supernatant. As a result, hybridomas that produce the desired antibodies having the ability to bind to the antigen can be cloned by limiting dilution method or a like. In this case, antigens that can preferably be used include not only the one that was used in immunization but also the GRP78 protein which is substantially of the same nature. [0070] Aside from the method of obtaining the above-mentioned hybridomas by immunizing animals other than humans with the antigen, human lymphocytes may be sensitized with the antigen to obtain the desired antibodies. Specifically, human lymphocytes are sensitized in vitro with the GRP78 protein. Subsequently, the immunosensitized lymphocytes are fused to a suitable fusion partner. An exemplary fusion partner that can be used is myeloma cells that derive from humans and which are capable of permanent division (see JP 1-59878 B). The anti-GRP78 antibody obtained by this method is a human antibody having an activity for binding to the GRP78 protein. [0071] Further, by administering the antigen GRP78 protein to a transgenic animal having the full repertoire of human antibody genes, the anti-GRP78 human antibody can also be obtained. The antibody producing cells in the immunized animal can be immortalized by such treatments as cell fusion with a suitable fusion partner and infection with Epstein-Barr virus or the like. From the thus obtained immortal cells, a human antibody against the GRP78 protein may be isolated (see WO 94/25585, WO 93/12227, WO 92/03918, and WO 94/02602). Further, the immortalized cells may be cloned to achieve cloning of cells that produce an antibody having the desired reaction specificity. If a transgenic animal is to be immunized, the immune system in that animal recognizes human GRP78 as foreign - 28 antigen. Hence, human antibodies against human GRP78 can be readily obtained. Hybridomas that produce the thus created monoclonal antibodies can be serially cultured in a conventional culture medium. If desired, those hybridomas can also be preserved in liquid nitrogen for an extended period. [0072] The hybridomas may be cultured in accordance with an ordinary method and the desired monoclonal antibodies may be obtained from the culture supernatant. Alternatively, the hybridomas may be administered to a compatible mammal and allowed to proliferate, yielding monoclonal antibodies in the ascites. The former method is suitable for obtaining antibodies of high purity. [0073] In the present invention, it is also possible to use antibodies that are encoded by the antibody gene cloned from the antibody producing cells. The cloned antibody gene may be incorporated into a suitable vector which is then introduced into a host cell so that it is expressed as an antibody. Methods for isolating the antibody gene, introducing it into a vector, and transforming host cells have already been established (see, for example, Vandamme, A. M. et al., Eur.J. Biochem. (1990) 192, 767-775). [0074] For example, cDNA coding for the variable region (V region) of the anti-GRP78 antibody can be obtained from hybridoma cells that produce the anti-GRP78 antibody. To this end, total RNA is typically first extracted from the hybridomas. Exemplary methods for extracting mRNA from cells include guanidine ultracentrifugation (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299) and the AGPC method (Chomczynski, P. et al., Anal. Biochem. (1987) 162, 156-159). [0075] The extracted mRNA can be purified using a mRNA Purification Kit (product of GE Healthcare Bioscience) or the like. Alternatively, kits for extracting total mRNA directly from cells are commercially available, as exemplified by a QuickPrep mRNA Purification Kit (product of GE Healthcare Bioscience). These kits may be used to obtain total mRNA from the hybridoma cells. From the obtained mRNA, cDNA coding for the V region of the antibody can be synthesized using a reverse transcriptase. It is also possible to synthesize cDNA by an AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (product of - 29 - SEIKAGAKU CORPORATION) and the like. In addition, for cDNA synthesis and amplification, one can utilize the 5'-RACE method (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 8998-9002; and Belyavsky, A. et al., Nucleic Acids Res. (1989) 17, 2919-2932) using 5'-Ampli FINDER RACE Kit (product of Clontech) and PCR. Furthermore, in this process of cDNA synthesis, a suitable restriction site (to be described later) may be introduced at both ends of cDNA. [0076] The desired cDNA fragment is purified from the obtained PCR product and subsequently ligated to a vector DNA. A recombinant vector is thusly constructed and introduced into E. coli or the like; after colony selection, a desired recombinant vector can be prepared from E. coli that has formed those colonies. Then, to determine whether the recombinant vector contains the nucleotide sequence of the desired cDNA, a known method such as the dideoxynucleotide chain termination technique may be adopted. [0077] To obtain a gene coding for the variable region, it is also possible to utilize a PCR technique that employs primers for amplifying the variable region gene. First, the extracted mRNA is used as a template to synthesize cDNA to make a cDNA library. To synthesize a cDNA library, a commercial kit is conveniently used. In practice, an extremely small amount of mRNA can be obtained from a small number of cells alone, so only low yield results from direct purification of the mRNA. Hence, purification is conventionally performed after adding a carrier mRNA that obviously contains no antibody gene. Alternatively, if a certain amount of RNA can be extracted, the RNA of the antibody producing cells suffices for efficient extraction. For instance, RNA extraction from at least 10 or at least 30, preferably at least 50 antibody producing cells sometimes requires no addition of a carrier RNA. [0078] The obtained cDNA library used as a template to amplify the antibody gene by a PCR method. Primers for amplifying the antibody gene by a PCR method are known. For example, primers for amplifying the human antibody gene can be designed on the basis of the disclosure in a scientific paper (J. Mol. Biol. (1991) 222, 581-597) or the like. These primers have different nucleotide sequences for different subclasses of immunoglobulin. Hence, if the cDNA library used as a template is unknown about which subclass it belongs to, every - 30 possibility must be considered in performing the PCR method. [0079] Specifically, if it is desired to acquire a gene coding for human IgG, the primers that can be utilized are those which enable amplification of genes coding for 71-75 heavy chains as well as K and X light chains. To amplify the variable region of IgG gene, a primer that anneals to a portion corresponding to the hinge region is typically used as the primer on the 3' side. On the other hand, a primer specific for the relevant subclass may be used as the primer on the 5' side. [0080] PCR products obtained by using primers for amplifying the genes for the respective subclasses of heavy and light chains are used as independent libraries. Utilizing the thus synthesized libraries, immunoglobulins comprising the combination of heavy and light chains can be reconstituted. The activities of the reconstituted immunoglobulins for binding to GRP78 may be used as an index for screening the desired antibodies. [0081] It is more preferred that the antibody of the present invention binds specifically to GRP78. Antibodies that bind to GRP78 can typically be screened through the following steps of: (1) contacting with GRP78 an antibody that contains a V region encoded by the cDNA obtained from the hybridomas; (2) detecting the binding between GRP78 and the antibody; and (3) selecting antibodies that bind to GRP78. [0082] Methods of detecting the binding between the antibody and GRP78 are known. Specifically, a subject antibody is reacted with GRP78 immobilized on a carrier, which is then treated with a labeled antibody that recognizes the antibody. Detection of the labeled antibody on the carrier after washing provides a proof for the binding of the subject antibody to GRP78. Labels that can be utilized include enzymatically active proteins such as peroxidase and p-galactosidase, and fluorescent substances such as FITC. To evaluate the binding activity of the antibody, fixed specimens of GRP78 expressing cells can also be utilized. [0083] An applicable method of screening antibodies using the binding activity as an index - 31 is panning that utilizes a phage vector. If the antibody gene is acquired as described above in a library of heavy and light chain subclasses, it is advantageous to perform screening utilizing a phage vector. Genes coding for the variable regions of heavy and light chains may be ligated by a suitable linker sequence to make a single-chain Fv (scFv). If the gene coding for scFv is inserted into a phage vector, phages can be obtained that have scFv expressed on the surface. These phages are brought into contact with the desired antigen and the phages that have bound to the antigen are recovered, whereupon one can recover the DNA that codes for scFv having the desired binding activity. By repeating this procedure as necessary, the scFv having the desired binding activity can be enriched. [0084] The polynucleotide that codes for the antibody in the present invention may encode the full length of the antibody or may encode part of the antibody. Part of the antibody refers to any portion of the antibody molecule. Hereinafter, the term "antibody fragment" may sometimes be used to indicate a part of the antibody. Preferred antibody fragments of the present invention contain a complementarity-determining region (CDR) of the antibody. More preferably, the antibody fragment according to the present invention contains all three CDRs that compose the variable region. [0085] After cDNA coding for the V region of the desired anti-GRP78 antibody is obtained, this cDNA is digested with restriction enzymes that recognize those restriction sites which have been inserted at both ends of the cDNA. Preferred restriction enzymes recognize and digest those nucleotide sequences which are less likely to appear in the nucleotide sequences that compose the antibody gene. Further, in order to insert a single copy of digested fragment into a vector in the correct orientation, restriction enzymes that impart sticky ends are preferred. The thus digested cDNA that codes for the V region of the anti-GRP78 antibody may be inserted into a suitable expression vector to thereby provide an antibody-expressing vector. In this case, a gene coding for the antibody's constant region (C region) and a gene coding for the aforementioned V region may be fused in frame to provide a chimeric antibody. The term "chimeric antibody" as used herein implies that the constant region derives from a different organism than the variable region. Hence, in addition to - 32 heterogeneous (e.g. mouse-human) chimeric antibodies, human-human homogeneous chimeric antibodies are also emcompassed by the chimeric antibodies according to the present invention. A chimeric antibody-expressing vector can also be constructed by inserting the aforementioned V region gene into an expression vector that already has a constant region. [0086] Specifically, an expression vector is provided that harbors DNA coding for a desired antibody's constant region (C region) and a sequence to be recognized by a restriction enzyme that digests the aforementioned V region gene may be located on the 5' side of the vector. The two genes are digested by the same combination of restriction enzymes and fused together in frame to construct a chimeric antibody-expressing vector. [0087] To produce the anti-GRP78 antibody of the present invention, the antibody gene may be incorporated into an expression vector in such a way that it will be expressed under control by an expression-regulatory region. The expression-regulatory region for expressing the antibody may include an enhancer or a promoter. Subsequently, suitable host cells are transformed with this expression vector to yield recombinant cells expressing the DNA coding for the anti-GRP78 antibody. [0088] Upon expressing the antibody genes, DNA coding for the antibody's heavy chain (H chain) and DNA coding for the light chain (L chain) may be respectively incorporated into different expression vectors. The vectors incorporating the H and L chains may be co-transfected in the same host cell to thereby express an antibody's molecule furnished with both H and L chains. Alternatively, DNA coding for the H chain and DNA coding for the L chain may be incorporated into a single expression vector for transforming host cells (see International Publication WO 94/11523). [0089] A lot of combinations of host and expression vector are known for first isolating the antibody gene and then introducing it into a suitable host to prepare an antibody. All of these expression systems can be applied in the present invention. In the case of using eukaryotic cells as a host, animal cells, plant cells or fungal cells may be used. Specifically, animal cells that may be employed in the present invention include, for example, mammalian cells (e.g. - 33 - CHO, COS, myeloma, BHK (baby hamster kidney), Hela, and Vero), amphibian cells (e.g. Xenopus oocytes), and insect cells (e.g. Sf9, Sf21, and Tn5). [0090] Alternatively, plant cells may be used and in this case, systems for expressing the antibody gene in cells of species derived from the genus Nicotiana such as Nicotiana tabacum are known. For transformation of plant cells, cells obtained by callus culture may be utilized. [0091] Furthermore, fungal cells that may be used include yeasts (the genus Saccharomyces including Saccharomyces serevisiaeand the genus Pichia including Pichiapastoris) and filamentous fungi (the genus Aspergillus including Aspergillus niger). [0092] Alternatively, expression systems are known for expressing the antibody gene utilizing prokaryotic cells. For example, in the case of using bacterial cells, E coli, Bacillus subtilis and other bacterial cells may be utilized in the present invention. [0093] In the case of using mammalian cells, an expression vector can be constructed that contains a commonly employed useful promoter, the antibody gene to be expressed, and a poly(A) signal that is functionally linked to the downstream of the 3' end. An exemplary promoter/enhancer is a human cytomegalovirus immediate early promoter/enhancer. [0094] Other types of promoter/enhancer that can be used to express the antibody of the present invention include a viral promoter/enhancer and a mammalian cell derived promoter/enhancer such as a human elongation factor-la (HEFla). Viruses that can utilize the promoter/enhancer may specifically be exemplified by retrovirus, polyoma virus, adenovirus, and simian virus 40 (SV40). [0095] In the case of using the SV40 promoter/enhancer, the method of Mulligan et al. (Nature (1979) 277, 108) may be utilized. In addition, the HEFla promoter/enhancer may be readily utilized by the method of Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322) to express the desired gene. [0096] In the case of E. coli, a commonly employed useful promoter, a signal sequence for antibody secretion and the antibody gene to be expressed may be functionally linked together to express the gene of interest. Exemplary promoters include a lacZ promoter and an araB - 34 promoter. In the cae of using a lacZ promoter, the method of Ward et al. (Nature (1989) 341, 544-546; FASEBJ. (1992) 6, 2422-2427) may be utilized. Alternatively, an araB promoter may be utilized to express the desired gene by the method of Better et al. (Science (1988) 240, 1041-1043). [0097] For antibody secretion, a pelB signal sequence (Lei, S. P. et al., J. Bacteriol. (1987) 169, 4379) may be used as a signal sequence in the case of producing the antibody in the periplasm of E coli. After separating the antibodies produced in the periplasm, a protein denaturing agent such as a guanidine hydrochloride of urea is used to thereby refold the structure of the antibodies in such a way that they have the desired binding activity. [0098] Replication origin to be inserted into the expression vector may be that derived from SV40, polyoma virus, adenovirus, and bovine papilloma virus (BPV). Further in addition, a selection marker may be inserted into the expression vector in order to increase the number of gene copies in the host cell system. Specifically, selection markers that may be utilized include an aminoglycoside transferase (APH) gene, a thymidine kinase (TK) gene, an E. coli xyanthine guanine phosphoribosyl transferase (Ecogpt) gene, a dihydrofolate reductase (dhfr) gene, etc. [0099] These expression vectors are introduced into host cells and the transformed host cells are cultured in vitro or in vivo to produce the desired antibody. The host cells are cultured in accordance with known methods. For example, DMEM, MEM, RPMI1640 or IMDM may be used as a culture medium, with a serum supplement such as fetal calf serum (FCS) being optionally used in combination. [0100] The antibodies expressed and produced as described above can be purified by the methods used to purify ordinary proteins and they may be used either singly or in suitable combinations. For example, an affinity column such as a protein A column, a chromatographic columm, a filter, ultrafiltration, salting out, dialysis, etc. may appropriately be selected and combined to separate and purify the antibodies (Antibodies-A Laboratory Manual, Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988). [0101] In addition to the above-mentioned host cells, transgenic animals can also be utilized - 35 to produce recombinant antibodies. To be more specific, such antibodies can be obtained from an animal into which a gene coding for the desired antibody has been introduced. For example, a fused gene containing the antibody gene can be constructed by inserting it in frame into a gene coding for the protein that is inherently produced in milk. An example of the protein to be secreted in milk is goat P casein. A DNA fragment containing the fused gene into which the antibody gene has been inserted is injected into a goat embryo, which is then introduced into a female goat. From the milk produced by a transgenic goat (or a offspring of the transgenic goat) born from the goat that received the embryo, the desired antibody can be acquired in the milk as a protein fused with the protein. In addition, in order to ensure that the milk containing the desired antibodies is produced from the transgenic goat in an increased amount, a hormone may be appropriately used in the transgenic goat (Ebert, K.M. et al., Bio/Technology (1994) 12, 699-702). A C region derived from an animal antibody can be used as the C region of the recombinant antibody of the present invention. For example, Cyl, Cy2a, Cy2b, Cy3, Cii, Cd, Cal, Ca2 or Ce may be used as the C region of H chains of the mouse antibody, and CK or C as the C region of L chains. In addition to the mouse antibody, applicable animal antibodies include those of rat, rabbit, goat, sheep, camel, monkey, etc. The sequences of these animal antibodies are known. In addition, in order to improve the stability of antibodies or of their production, the C region may be modified. In the case where antibodies are to be administered to humans in the present invention, artificially modified recombinant antibodies may be employed for purposes of reducing a heterogenic antigenicity to humans. Examples of the recombinant antibodies include a chimeric antibody and a humanized antibody. [0102] These modified antibodies can be produced using known methods. Chimeric antibodies are such that a variable region and a constant region that derive from different sources from each other are linked together. For example, an antibody comprising variable regions of heavy and light chains of a mouse antibody and constant regions of heavy and light chains in a human antibody is a mouse-human heterogeneous chimeric antibody. The DNA coding for the variable regions of the mouse antibody is linked to the DNA coding for - 36 the constant regions of the human antibody and the linked DNAs are incorporated into an expression vector to construct a recombinant vector that expresses a chimeric antibody. Recombinant cells transformed with the recombinant vector are cultured to express the incorporated DNAs, whereby the chimeric antibodies can be acquired as they are produced during culture. The C regions of the human antibody are used as those of the chimeric antibody and the humanized antibody. For example, in H chains, Cyl, Cy2, Cy3, Cy4, Cli, Cd, Cal, Ca2 and Ce can be utilized as C regions. In L chains, CK and C), can be used as C regions. The amino acid sequences of these C regions and the nucleotide sequences encoding thereof are known. In addition, in order to improve the stability of antibodies per se or of production of antibodies, the C regions of the human antibody may be modified. [0103] In general, chimeric antibodies are composed of V regions of an antibody derived from non-human animals and C regions derived from a human antibody. In contrast, humanized antibodies are composed of a complementarity-determining region (CDR) of an antibody derived from non-human animals, a framework region (FR) derived from a human antibody, and a C region derived from a human antibody. The humanized antibody has lowered antigenicity in the human body, so it is useful as an active ingredient in the therapeutics of the present invention. [0104] The variable region of an antibody is typically composed of three complementarity determining regions (CDRs), each located between four frame regions (FRs). CDRs are regions that substantially determine the binding specificity of the antibody. The amino acid sequences of CDRs are highly diverse. On the other hand, the amino acid sequences that compose FRs often show high homology between antibodies even if they have different binding specificities. Hence, it is generally held that by translocating CDRs, the binding specificity of a certain antibody can be translocated into another antibody. [0105] The humanized antibody is also called a reshaped human antibody. Specifically, a humanized antibody obtained by translocating CDRs of an antibody from a non-human animal, such as mouse, into the human antibody is known. General recombinant techniques for obtaining humanized antibodies are also known. -37- [0106] Specifically, overlap extension PCR is known as a method of translocating CDRs of a mouse antibody into human FRs. In overlap extension PCR, nucleotide sequences that encode CDRs of the mouse antibody to be translocated are attached to primers for synthesizing the FRs of a human antibody. A primer is prepared for each of the four FRs. It is generally considered that when translocating mouse CDRs into human FRs, selecting human FRs with high homology to the mouse FRs is advantageous by selected for the purpose of maintaining the function of CDRs. In other words, it is generally preferred to utilize human FRs that are composed of amino acid sequences having high homology to the amino acid sequences of FRs adjacent to the mouse CDRs to be translocated. [0107] The nucleotide sequences to be linked are so designed that they will be linked together in frame. Human FRs are individually synthesized by the respective primers. As a result, there are obtained products in which DNAs coding for the mouse CDRs are attached to the respective FRs. The nucleotide sequences coding for the mouse CDRs in the respective products are so designed that they overlap each other. Subsequently, with the human antibody gene used as a template, the overlapped CDR regions of the synthesized products are annealed to each other to perform a complementary strand synthesis reaction. This reaction causes the human FRs to be linked together via the sequences of mouse CDRs. [0108] Finally, full length of the V region gene in which three CDRs are linked to four FRs are amplified by means of primers that are annealed to its 5' and 3' ends and which have suitable restriction enzyme recognition sequences added thereto. The thus obtained DNA and the DNA coding for the C region of the human antibody are inserted into an expression vector such that they will be fused together in frame, whereby a humanized antibody expression vector can be constructed. This recombinant vector is introduced into host cells to establish recombinant cells, which are then cultured to express the DNA coding for the humanized antibody, whereupon the humanized antibodies are produced in the culture of the cultured cells (see European Patent Publication EP 239400 and International Publication WO 96/02576). [0109] The thus prepared humanized antibodies are qualitatively or quantitatively measured - 38 and evaluated for their activity for binding to antigens and this allows for advantageous selection of those FRs of the human antibody which can be linked together via CDRs such that the CDRs will form desired antigen binding sites. If necessary, the amino acid residues of FRs may be replaced such that the CDRs of the reconstructed human antibody will form appropriate antigen binding sites. For instance, alterations of the amino acid sequences can be introduced into FRs by applying the PCR method used to translocate the mouse CDRs into the human FRs. Specifically, alterations of partial nucleotide sequences can be introduced into the primers that are to anneal to the FRs. The FRs synthesized by means of such primers have the variations in nucleotide sequences introduced thereinto. The variant antibody with substituted amino acids is measured and evaluated for its binding activity to antigens by the above-described methods, whereupon variant FR sequences having the desired properties can be selected (Sato, K. et al., Cancer Res, 1993, 53, 851-856). [0110] Methods of acquiring the human antibody are also known in the art. For instance, human lymphocytes are sensitized in vitro with desired antigen or cells expressing the desired antigen. Subsequently, the sensitized lymphocytes are fused to human myeloma cells, whereby the desired human antibodies can be acquired that have the activity for binding to the antigen (see JP 1-59878 B). The human myeloma cells that are used as the fusion partner may be exemplified by U266. [0111] The desired human antibody can also be acquired by immunizing with the desired antigen a transgenic animal having the full repertoire of human antibody genes (see International Publications WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, and WO 96/33735). Also known is a technique that uses panning method with a human antibody library to acquire the human antibody. For example, the V region of a human antibody can be expressed as a single-chain antibody (scFv) on the surface of phages by the phage display method and those phages that bind to the antibody can be selected. By analyzing the gene of the selected phages, the DNA sequence that codes for the V region of the human antibody that binds to the antigen can be determined. After determining the DNA sequence of scFv that binds to the antigen, this V region sequence is -39fused in frame to the sequence of the desired human antibody's C region, which is then thereafter inserted into a suitable expression vector to construct expression vectors. The expression vectors are then introduced into the suitable host cells mentioned above so that the gene coding for the desired human antibody is expressed in the host cells to yield the desired human antibodies. These methods are already known (International Publications WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438 and WO 95/15388). [0112] As long as they bind to the GRP78 protein, the antibodies of the present invention include not only bivalent antibodies such as IgG but also monovalent antibodies, as well as polyvalent antibodies such as IgM. The polyvalent antibodies according to the present invention include those whose antigen binding sites are identical, as well as those having partly or entirely different antigen binding sites. The antibodies of the present invention are not limited to full-length molecules of antibodies and as long as they bind to the GRP78 protein, they may be small molecular antibodies or their modifications. [0113] The small molecular antibodies include antibody fragments, or fragments of a whole antibody (e.g. whole IgG) which are deficient of partial regions. As long as the ability to bind to the GRP78 antigen is retained, the partially deficient antibody molecule is available. The antibody fragment in the present invention preferably contains either a heavy-chain variable region (VH) or a light-chain variable region (VL) or both. The amino acid sequence of VH or VL may contain a substitution, deletion, addition and/or insertion. Furthermore, as long as the ability to bind to the GRP78 antigen is retained, either VH or VL or a part of both regions may be deficient. If desired, the variable region may be chimeric or humanized. Specific examples of the antibody fragment include Fab, Fab', F(ab')2 and Fv. Specific examples of the small molecular antibody include Fab, Fab', F(ab')2, Fv, scFv (single-chain Fv), Diabody, and sc(Fv)2 (single-chain (Fv)2). Multimers of these antibodies (e.g. dimers, trimers, tetramers and polymers) are also encompassed by the small molecular antibodies according to the present invention. [0114] Fragments of antibodies can be obtained by treating the antibodies with enzymes to - 40 generate antibody fragments. Known examples of enzymes that can generate antibody fragments include papain, pepsin, and plasmin. Alternatively, a gene coding for one of these antibody fragments may be constructed, introduced into an expression vector, and expressed in a suitable host cell (see, for example, Co, M.S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. & Horwitz, A. H. Methods in Enzymology (1989) 178, 476-496; Plueckthun, A. & Skerra, A. Methods in Enzymology (1989) 178, 476-496; Lamoyi, E., Methods in Enzymology (1989) 121, 652-663; Rousseaux, J. et al., Methods in Enzymology (1989) 121, 663-669; Bird, R. E. et al., TIBTECH (1991) 9, 132-137). [0115] Digesting enzymes cleave antibody fragments at specific positions to provide antibody fragmens having the following specific structures. Such enzymatically obtained antibody fragments may further be treated by genetic engineering techniques to delete any desired portions of the antibody. Papain digestion: F(ab)2 or Fab; Pepsin digestion: F(ab')2 or Fab'; Plasmin digestion: Facb. [0116] The "diabody" refers to a bivalent antibody fragment constructed by gene fusion (Holliger P et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP 404097, WO 93/11161, etc.). A diabody is a dimer composed of two polypeptide chains. Each of the polypeptide chains that compose a dimer is usually such that VL and VH are linked by a linker in the single chain. The linkers in a diabody are typically so short that VL and VH in the single chain away from the linker cannot bind to each other. Specifically, the amino acid residues that compose a linker may be about five in number. Hence, VL and VH that are encoded on the single polypeptide chain cannot form a single-chain variable region fragment but forms a dimer with another single-chain variable region fragment. As a result, the diabody will have two antigen binding sites. [0117] The scFv is obtained by linking the V region of H chain and the V region of L chain in the antibody. In scFv, the V region of H chain and the V region of L chain are linked via a linker, preferably a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A, 1988, 85, - 41 - 5879-5883). The V region of H chain and the V region of L chain in scFv may be derived from any of the antibodies mentioned herein. The peptide linker that links the two V regions is not particularly limited. For example, any single-chain peptide composed of from about 3 to 25 amino acid residues may be used as a linker. The V regions can typically be linked by the PCR method described above. For linking the V regions by the PCR method, DNA coding for the amino acid sequences of the DNA sequence coding for the H chain or its V region in the aforementioned antibody and of the DNA sequence coding for the L chain or its V region in the aforementioned antibody is utilized as a template. [0118] By the PCR method using a pair of primers that have sequences corresponding to the sequences at both ends of the DNA to be amplified, the DNA coding for the V region of H chain and the DNA coding for the V region of L chain are each amplified. Subsequently, DNA coding for the peptide linker portion is prepared. The DNA coding for the peptide linker can also be synthesized by utilizing PCR. In this instance, a separately synthesized an additional nucleotide sequence that can link to an amplified product of each V region is attached to the 5' end of each of the primers to be utilized. Subsequently, a PCR reaction is performed utilizing each of the DNAs of [H-chain V region DNA]-[peptide linker DNA]-[L chain V region DNA] and primers for assembly PCR. The primers for assembly PCR comprise the combination of a primer that anneals to the 5' end of [H-chain V region DNA] and a primer that anneals to the 3' end of [L-chain V region DNA]. In other words, the primers for assembly PCR constitute a primer set that can amplify the DNA coding for the full-length sequence of scFv to be synthesized. On the other hand, a nucleotide sequence that can link to the DNA of each V region is attached to [peptide linker DNA]. As a result, the DNAs described above are linked and with the aid of the primers for assembly PCR, the full length of scFv is eventually generated as an amplified product. Once the DNA coding for scFv is constructed, an expression vector that contains it and recombinant cells transformed with that expression vector can be acquired in accordance with the usual manner. In addition, the resulting recombinant cells may be cultured to express the DNA coding for the scFv, whereupon the scFv can be acquired. - 42 - [0119] Note that sc(Fv)2 is a small molecular antibody obtained by binding two VHs and two VLs into a single chain with linkers or the like (Hudson et al, J Immunol. Methods 1999; 231: 177-189). This sc(Fv)2 can typically be prepared by connecting two scFv molecules with a linker. [0120] A preferred antibody is such that two VHs and two VLs are arranged in the order of VH, VL, VH and VL (i.e., [VH]-linker-[VL]-linker-[VH]-linker-[VL]), starting at the N-terminal side of a single-chain polypeptide. [0121] The order of two VHs and two VLs is by no means limited to the arrangement shown above and they may be arranged in any other orders, including the following arrangements: [VL] -linker- [VH]-linker- [VH] -linker- [VL]; [VH] -linker- [VL]-linker-[VL] -linker-[VH]; [VH] -linker- [VH] -linker- [VL]-linker-[VL]; [VL]-linker-[VL]-linker-[VH]-linker-[VH]; and [VL] -linker- [VH] -linker- [VL] -linker- [VH]. [0122] Linkers that may be used to link the variable regions of an antibody include any peptide linkers that can be introduced by genetic engineering and synthetic compound linkers (e.g. the linker disclosed in Protein Engineering, 9(3), 299-305, 1996). Peptide linkers are preferred in the present invention. The length of peptide linkers is not particularly limited and may be selected by skilled artisans as appropriate for a specific object. The amino acid residues that compose peptide linkers typically consist of 1 to 100 amino acids, preferably 3 to 50 amino acids, more preferably 5 to 30 amino acids, and most preferably 12 to 18 amino acids (say, 15 amino acids). [0123] The peptide linkers may be composed of any amino acid sequences as long as they will not compromise the binding action of scFv. [0124] Alternatively, synthetic compound linkers (chemical crosslinking agents) may also be utilized to link the V regions. Crosslinking agents commonly used to crosslink peptide compounds and the like may be utilized in the present invention. Examples that may be used - 43 include N-hydroxysuccinimide (NHS), disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl)suberate (BS3), dithiobis(succinimidyl propionate) (DSP), dithiobis(sulfosuccinimidyl propionate) (DTSSP), ethylene glycol bis(succinimidyl succinate) (EGS), ethylene glycol bis(sulfosuccinimidyl succinate) (sulfo-EGS), disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), bis[ 2 -(succinimidooxycarbonyloxy)ethyl]sulfone (BSOCOES), and bis[ 2 -(sulfosuccinimidooxycarbonyloxy)ethyl]sulfone (sulfo-BSOCOES). [0125] To link four variable regions of an antibody together, three linkers are usually required. The linkers may be the same or different from each other. A small molecular antibody that is preferred in the present invention is a diabody or sc(Fv)2. To obtain such small molecular antibodies, an antibody may be treated with an enzyme such as papain or pepsin to generate antibody fragments; alternatively, DNA coding for such antibody fragments may be constructed, introduced into an expression vector, and expressed in a suitable host cell (see, for example, (Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, A. H., Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E., Methods Enzymol. (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol. (1986) 121, 663-669; Bird, R. E. and Walker, B. W., Trends Biotechnol. (1991) 9, 132-137). [0126] Further, the antibody of the present invention may be used as a modified antibody in which it is bound to various molecules such as polyethylene glycol (PEG). Such modified antibodies can be obtained by chemically modifying the antibody of the present invention. Methods of modifying antibodies have already been established in the art. [0127] Further in addition, the antibody of the present invention may be a bispecific antibody. The bispecific antibody refers to such an antibody that variable regions that recognize different epitopes are present in the same antibody molecule; the epitopes may be present in different molecules or they may be present in the same molecule. Thus, in the present invention, the bispecific antibody may possess antigen binding sites that recognize different epitopes on the GRP78 molecule; alternatively, it may be such a bispecific antibody - 44 that one of the two recognition sites recognizes GRP78 whereas the other recognizes a cytotoxic substance. The "antibody" in the present invention encompasses these antibodies. [0128] In the present invention, the antibody may be constructed by combining with bispecific antibodies that recognize antigens other than GRP78. For example, it may be combined with a bispecific antibody that recognizes an antigen different from GRP78 but which, like GRP78, is specifically expressed on the cell surface of the target cancer cells. [0129] Methods for producing bispecific antibodies are known. For example, two kinds of antibody that recognize different antigens may be bound to construct a bispecific antibody. Each of the antibodies to be bound may be a 1/2 molecule that has both H and L chains or it may be a 1/4 molecule having only a H chain. Alternatively, hybridomas producing different monoclonal antibodies may be fused to construct bispecific antibody producing fused cells. Still another way to prepare a bispecific antibody is by genetic engineering techniques. Binding Activity of Antibody [0130] The activity of antibodies for binding to antigens can be measured by known methods (Antibodies - A Laboratory Manual. Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988). For example, ELISA (enzyme-linked immunosorbent assay), EIA (enzyme immunoassay), RIA (radioimmunoassay) or immunofluorescence may be employed. Further, techniques for measuring the activity of antibodies for binding to antigens expressed in cells include the method described in Antibodies - A Laboratory Manual, ibid, pp. 359-420. [0131] To measure the binding between an antigen expressed on the surface of cells suspended in a buffer solution or the like and the antibody against that antigen, a method using a flow cytometer may advantageously be employed. Examples of the flow cytometr TM TM TM that may be used include FACSCantol" II, FACSAria , FACSArray TM, FACSVantage SE, and FACSCalibur TM (these are available from BD Biosciences), as well as EPICS ALTRA HyPerSort, Cytomics FC 500, EPICS XL-MCL ADC EPICS XL ADC, and Cell Lab Quanta/Cell Lab Quanta SC (these are available from Beckman Coulter). [0132] An advantgageous method for measuring the activity of the GRP78 antibody to bind to antigens comprises a method comprising steps of reacting the subject antibody with cells - 45 expressing GRP78, staining the subject antibody with an FITC-labeled secondary antibody that recognizes the subject antibody, performing measurement with FACSCalibur (BD Biosciences), and analyzing the fluorescence intensity data with CELL QUEST Software (BD Biosciences). Growth Supressing Activity [0133] To evaluate or measure the cell growth suppressing effect based on the anti-GRP78 antibody, the following methods may advantageously be employed. A method for evaluating or measuring this cell growth suppressing activity in vitro is such that the uptake by viable cells of [ 3 H]-labeled thymidine added into a culture medium is measured as an index for DNA replicating ability. More convenient methods that may be employed include a dye exclusion method in which the ability to exclude a dye such as trypan blue to the outside of cells is measured under a microscope, as well as the MTT method. The latter method utilizes the ability of viable cells to convert the tetrazolium salt MTT (3-(4,5-dimethylthiazol-2-yl) 2,5-diphenyl tetrazolium bromide) to a blue formazan product. More specifically, the subject antibody as well as a ligand is added to a culture medium containing the subject cells and after a certain period of time, an MTT solution is added to the culture medium, which is allowed to stand for a certain period of time, whereupon MTT is incorporated into the cells. As a result, the MTT which is a yellow compound is converted to a blue compound by means of succinate dehydrogenase in the intracellular mitochondria. The blue product is dissolved to develop color, whose absorbance is measured for use as an index for the viable cell count. Aside from MTT, reagents such as MTS, XTT, WST-1 and WST-8 are also available on the market (as from nacalai tesque) and may advantageously be used. For activity measurement, a control antibody may also be used. [0134] A tumor bearing mouse model may be used as a method of in vivo evaluation or measurement of the cell growth suppressing activity. For example, cancer cells expressing GRP78 are transplanted intradermally or subcutaneously into a subject non-human animal and starting on the same day or the next day, the subject antibody is administered intravenously or intraperitoneally every day or at intervals of several days. The size of the - 46 tumor is measured over time (in days) to evaluate the cell growth suppressing activity of the subject antibody. As in the evaluation in vitro, a control antibody is administered and if the tumors in the group administered with the anti-GRP78 antibody are significantly smaller than the tumors in the group administered with the control antibody, the subject antibody may be found to have the cell growth suppressing activity. If a mouse is used as a subject non-human animal, a nude (nu/nu) mouse that hereditarily lacks the thymus to be deprived of the function of T lymphocytes may advantageously be used. By using this mouse, the involvement of T lymphocytes in the subject animal can be eliminated when the cell growth suppressing activity of the administered antibody is evaluated or measured. Method of Supressing the Growth of Cells [0135] The present invention provides a method of suppressing the growth of GRP78 expressing cells by bringing them into contact with the antibody of the present invention. The antibody of the present invention has already been described on the foregoing pages as an antibody that binds to the GRP78 protein contained in the cell growth suppressing agent of the present invention. The cells to be contacted by the anti-GRP78 antibody are not particularly limited if GRP78 has been expressed; preferably, they are such cells that GRP78 is localized on the cell membrane, and they are preferably associated with disease. Preferred examples of cells associated with disease include cancer cells. Also included are vascular endothelial cells that are present in a malignant tumor (tumor vessels) in the preferred examples of cells. The target cancer types are not particularly limited and include, for example, prostate cancer, breast cancer, pancreas cancer, liver cancer, lung cancer, esophagus cancer, melanoma, colon cancer, stomach cancer, ovary cancer, bladder cancer, and brain tumor. Method of Delivery Using Anti-GRP78 Antibody [0136] The present invention relates to a method of delivering a cytotoxic substance into cells using the -GRP78 antibody. The antibody to be used in the present invention is the anti-GRP78 antibody that is conjugated with the above-mentioned cytotoxic substance. In this case, it is preferably an antibody having an internalizing activity. Delivery of the - 47 cytotoxic substance can be performed by establishing contact between the anti-GRP78 antibody that is conjugated with the cytotoxic substance and a cell that expresses GRP78. In the present invention, the cells into which the cytotoxic substance is to be delivered are not particularly limited but they are preferably such cells that GRP78 is localized on the cell membrane, and they are preferably associated with disease. Preferred examples of cells associated with disease include cancer cells. Also included are vascular endothelial cells that are present in a malignant tumor (tumor vessels) in the preferred examples of cells. The target cancer types are not particularly limited and include, for example, prostate cancer, breast cancer, pancreas cancer, liver cancer, lung cancer, esophagus cancer, melanoma, colon cancer, stomach cancer, ovary cancer, bladder cancer, and brain tumor. [0137] In the present invention, the contact may be established in vitro or in vivo. In these cases, the antibody to be added may appropriately be in the form of a solution or of solid which is formed by freeze-drying or otherwise in other forms. If the antibody is to be added in an aqueous solution, the aqueous solution may contain only the antibody as the sole component or the antibody with other substances such as a surfactant, an excipient, a coloring agent, an odorizer, a preservative, a stabilizer, a buffer, a suspending agent, an isotonization agent, a binder, a disintegrator, a lubricant, a fluidity promoter, a flavoring agent, etc. The concentration at which the antibody is added is not particularly limited and it may advantageously be used to give a final concentration in the culture medium which is preferably in the range from 1 pg/ml to 1 g/ml, more preferably from 1 ng/ml to 1 mg/ml, and even more preferably from 1[tg/ml to 1 mg/ml. [0138] In the present invention, in vivo "contact" may also be effected by administration into a non-human animal which has GRP78 expressing cells transplanted into their body or into animals, including humans, that possess cancer cells expressing GRP78 endogenously. Administration can be performed either orally or parenterally. Administration by parenteral route is particularly preferred and specific examples of this method include injection, transnasal administration, transpulmonary administration, and transdermal administration. In the case of an injection, the pharmaceutical composition, the cell growth inhibitor and the - 48 anticancer agent according to the present invention can be administered systemically or topically by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection or the like. In addition, the method of administration can appropriately be selected for the age or symptoms of the subject animal. If the antibody is administerred in the form of an aqueous solution, the aqueous solution may contain only the antibody as the sole component or the antibody with other substances such as a surfactant, an excipient, a coloring agent, an odorizer, a preservative, a stabilizer, a buffer, a suspending agent, an isotonization agent, a binder, a disintegrator, a lubricant, a fluidity promoter, a flavoring agent, etc. The dosage may be selected from the range of 0.0001 mg to 1000 mg per kg of body weight for a single administration. Alternatively, the dosage can be selected from the range of 0.001 to 100,000 mg/body per patient. It should, however, be noted that the dosage of the antibody to be administered in the present invention is by no means limited to the values indicated above. Pharmaceutical Compositions [0139] In another aspect, the present invention is characterized by providing a pharmaceutical composition comprising an antibody that binds to the GRP78 protein. The present invention is also characterized by providing a cell growth suppressing agent, in particular, an anticancer agent that contains an antibody that binds to the GRP78 protein. The cell growth suppressing agent and anticancer agent of the present invention are preferably administgered to a subject who suffers or is suspected of suffering from cancer. [0140] In the present invention, the cell growth suppressing agent that contains an antibody that binds to the GRP78 protein may also be expressed as a method of suppressing cell growth which includes the step of administering a subject with an antibody that binds to the GRP78 protein or as use of such antibody in the manufacture of a cell growth suppressing agent. [0141] In the present invention, the anticancer agent that contains an antibody that binds to the GRP78 protein may be rewritten as a method of preventing or treating cancer which includes the step of administering a subject with an antibody that binds to the GRP78 protein - 49 or as use of such antibody in the manufacture of an anticancer agent. [0142] The antibody to be contained in the pharmaceutical composition (e.g. cell growth suppressing agent or anticancer agent) of the present invention is not particularly limited as long as it binds to the GRP78 protein, and any of the antibodies that are given as examples herein may be used. [0143] The pharmaceutical compositions of the present invention can be administered either orally or parenterally. Administration by parenteral route is particularly preferred and specific examples of this method include injection, transnasal administration, transpulmonary administration, and transdermal administration. In the case of an injection, the pharmaceutical compositions of the present invention can be administered systemically or topically by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection or the like. In addition, the method of administration can be selected as appropriate for the age or symptoms of the patient. The dosage may be selected from the range of 0.0001 mg to 1000 mg per kg of body weight for a single administration. Alternatively, the dosage can be selected from the range of 0.001 to 100,000 mg/body per patient. It should, however, be noted that the dosage of the pharmaceutical compositions to be administered in the present invention is by no means limited to the values indicated above. [0144] The pharmaceutical compositions of the present invention can be formulated according to the usual procedures (see, for example, Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A), optionally together with pharmaceutically acceptable carriers and additives. Examples include a surfactant, an excipient, a coloring agent, an odorizer, a preservative, a stabilizer, a buffer, a suspending agent, an isotonization agent, a binder, a disintegrator, a lubricant, a fluidity promoter, a flavoring agent, etc. but these are not the sole cases and other common carriers may be used as appropriate. Specific examples include light silicic anhydride, lactose, microcrystalline cellulose, mannitol, starch, carmelose calcium, carmelose sodium, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinyl acetal diethyl aminoacetate, polyvinyl pyrrolidone, gelatin, medium-chain fatty acid triglycerides, polyoxyethylene-hardened castor oil 60, - 50 sucrose, carboxymethylcellulose, corn starch, and inorganic salts. Process for Producing Pharmaceuticals [0145] The present invention further provides a process for producing a pharmaceutical, in particular, an anticancer agent, which comprises the following steps of: (a) providing anti-GRP78 antibodies; (b) validating whether the antibodies of (a) have an internalizing activity; (c) selecting antibodies having an internalizing activity; and (d) binding a cytotoxic substance to the antibodies selected in (c). [0146] The presence or absence of an internalizing activity can be confirmed by the methods described on the foregoing pages. As regards the anti-GRP78 antibodies and the cytotoxic substance, the anti-GRP78 antibody and cytotoxic substance that have been described on the foregoing pages may be employed. Diagnosis of Cancer [0147] The present invention also provides a method of diagnosing disease, in particular cancer, using an anti-GRP78 antibody. [0148] The diagnostic method of the present invention can be performed by detecting the anti-GRP78 antibody incorporated into cells. The anti-GRP78 antibody to be used in the present invention preferably has an internalizing activity, and it is also preferred that the antibody is labeled with a labeling substance. [0149] Hence, a preferred embodiment of the diagnostic method of the present invention is by using an anti-GRP78 antibody that is labeled with a labeling substance and which has an internalizing activity. The anti-GRP antibody to which a labeling substance is bound may be the anti-GRP78 antibody that has been described on the foregoing pages. [0150] The labeling substance to be bound to the anti-GRP78 antibody is not particularly limited and labeling substances known to skilled artisans such as a fluorescent dye, an enzyme, a co-enzyme, a chemoluminescent material, and a radioactive material may be employed; specific examples include radioisotopes (e.g. 32P, 14C, 1251, 3H, and 1311), fluorescein, rhodamine, dansyl chloride, umberiferron, luciferase, peroxidase, alkali - 51 phosphatase, -galactosidase, p-glucosidase, horse raddish peroxidase, glucoamylase, lysozyme, saccharide oxidase, microperoxidase, and biotin. If biotin is used as a labeling substance, after the addition of the biotin-labeled antibody, avidin that is conjugated with an enzyme, such as alkali phosphatase, is preferably added. To bind the labeling substance and the anti-GRP78 antibody, a known method may be employed, such as the glutaraldehyde method, maleimide method, pyridyl disulfide method, or the periodic acid method. [0151] The labeling substance can be conjugated with the antibody by any methods known to skilled artisans. [0152] In the case where the disease to be diagnosed by the method of the present invention is cancer, the target cancer types are not particularly limited and include, for example, prostate cancer, breast cancer, pancreas cancer, liver cancer, lung cancer, esophagus cancer, melanoma, colon cancer, stomach cancer, ovary cancer, bladder cancer, and brain tumor. [0153] Diagnosis in the present invention may be performed eiher in vivo or in vitro. [0154] If diagnosis is to be performed in vitro, diagnosis is conducted by a method comprising the following steps of: (a) providing a sample collected from a subject to be diagnosed; (b) bringing the sample of (a) to an anti-GRP78 antibody that is conjugated with a labeling substance; and (c) detecting antibodies that have been incorporated into cells. [0155] The sample to be collected is not particularly limited and cells or tissues collected from the subject to be diagnosed may be mentioned as examples. In addition, secondary samples obtained from the subject sample, such as specimens in which tissues or cells collected from the body of an organism have been fixed or a culture medium containing cells, are also included in the category of the sample according to the present invention. [0156] If diagnosis is to be performed in vivo, diagnosis is conducted by a method comprising the following steps of: (a) administering a subject to be diagnosed with an anti-GRP78 antibody that is conjugated with a labeling substance; and - 52 - (b) detecting antibodies that have been incorporated into cancer cells. [0157] The dosage of the anti-GRP78 antibody can appropriately be determined by skilled artisans based on the type of the labeling substance and the type of the disease to be diagnosed. The labeled anti-GRP78 antibody may be formulated by the methods described on the foregoing pages. [0158] The present invention further provides a process for producing a diagnostic agent, in particular one for cancer diagnosis, which comprises the following steps of: (a) providing anti-GRP78 antibodies; (b) validating whether the antibodies of (a) have an internalizing activity; (c) selecting antibodies having an internalizing activity; and (d) binding a labeling substance to the antibodies selected in (c). [0159] The presence or absence of an internalizing activity can be confirmed by the methods described on the foregoing pages. As regards the anti-GRP78 antibodies and the labeling substance, the anti-GRP78 antibody and labeling substance that have been described on the foregoing pages may be employed. Partial Peptides of GRP78 [0160] The present invention provides a polypeptide comprising the amino acid sequence of SEQ ID NO: 3 (the 3 7 6 th to 4 1 5 th of GRP78) or a fragment thereof. A polypeptide comprising the amino acid sequence of SEQ ID NO: 3 (the 3 7 6 th to 4 15 th of GRP78) or a fragment thereof is useful for evaluating the immunogen used in generating the antibody of the present invention or the binding activity of the antibody generated. In the present invention, fragments comprise at least 5 amino acids, preferasbly at least 10 amino acids, more preferably at least 15 amino acids. Examples of the fragments of polypeptides comprising the amino acid sequence of SEQ ID NO: 3 are not particularly limited and may include a fragment comprising the amino acid sequence from the 3 8 4 th to 3 9 1 s, of GRP78 (a fragment comprising the sequence of amino acids 9-16 in SEQ ID NO: 3), a fragment comprising the amino acid sequence from the 3 9 2 "d to 4 0 7 th of GRP78 (a fragment consisting of the sequence of amino acids 17-32 in SEQ ID NO: 3), and a fragment comprising the - 53 amino acid sequence from the 4 0 0 th to 4 15 th of GRP78 (a fragment comprising the sequence of amino acids 25-40 in SEQ ID NO: 3). EXAMPLES [0161] Example 1: Immunization 1-1. Preparing an immunogen 1-1-11. Constructing a GRP78 E. coli expressing vector [0162] To construct a GRP78 E. coli expressing vector, cloning of the GRP78 gene was first performed as follows. In the first place, with human colon adenocarcinoma cDNA (MTC Multiple Tissue cDNA panel, Clontech) used as a template, a full-length GRP78 gene was cloned by performing RT-PCR under the following conditions using Pyrobest Taq polymerase (Takara). GRP-1: atgaagctct ccctggtggc (SEQ ID NO: 26) GRP-2: ctacaactca tctttttctg ctgta (SEQ ID NO: 27) (94 *C x 30 sec, 58 *C x 30 sec, 72 C x 120 sec: 27 cycles) [0163] In the next step, with the obtained PCR product used as a template, another PCR was run under the following conditions to generate a GRP78 cDNA fragment having a BamHI and an XhoI cleavage sequence attached to the 5' and 3' ends, respectively, of a GRP78 gene fragment comprising base Nos. of 55-1965 in the nucleotide sequence of SEQ ID NO: 1. GRP-GST-1: aaaggatccg aggaggagga caagaaggag gacgtggg (SEQ ID NO: 28) GRP-GST-2: tttctcgagc tacaactcat ctttttctgc tgtatcctc (SEQ ID NO: 29) (94 'C x 30 sec, 64 C x 30 sec, 72 C x 120 sec: 25 cycles) [0164] The obtained GRP78 cDNA fragment was cleaved with BamHI and XhoI and inserted downstream of the GST code region of an E. coli GST fused expression vector that had been similarly cleaved with BamHI and XhoI (pGEX-6P-1, Amersham Pharmacia) to construct a GRP78-GST fused protein expression vector (pGEX-GRP78-full). 1-1-2. Inducing the expression of GST fused GRP78 protein and its purification [0165] In the next step, the GRP78 protein was prepared as an immunogen for acquiring - 54 - GRP78 binding antibodies. [0166] First, E coli (BL21) was transformed with PGEX-GRP78-full. The transformed cells were cultured in an LB medium (300 ML) and when an OD 6 10 value of at least 0.5 was reached, IPTG was added to give a concentration of 0.5 mM, whereby induction of protein expression was effected. After 5-hr culture, the E coli cells were harvested by centrifugation. [0167] The harvested E coli cells were suspended in 30 ml of B-PER (PIERCE) to be lysed. The E. coli lysate obtained was diluted 10-fold with PBS; to the diluted lysate, PBS-equilibrated glutathione Sepharose 4B (Amersham Pharmacia) was added, followed by incubation at 4 'C overnight. Thereafter, several washings with PBS were done to remove any unadsorbed proteins and reaction with PreScissuion Protease (Amersham Pharmacia) was performed at 4 'C overnight in a protease reaction solution (50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 1 mM DTT, pH 7.5). By this procedure, the GST protein and GRP78 protein (amino acids 19-654) in the GRP78-GST fused proein were separated off from each other. Subsequently, the GRP78 protein eluted by protease digestion was separated by gel filtration chromatography on Superdex 200 HR 10 30 column (Amersham Pharmacia) to recover the desired GRP78 protein (amino acids 19-654). 1-2. Immunization [0168] Emulsions of the GRP78 protein were prepared using COMPLETE ADJUVANT (DIFCO: DF263810) for priming and IMCOMPLETE ADJUVANT (DIFCO: DF263910) for booster; each of the emulsions was administered to three mice in the following groups [(MRL/lpr, male, 4-wk old) (Balb/c, female, 6-wk old): all purchased from charles river, Japan] at 50 kg/mouse by subcutaneous injection to immunize them (TERUMO syringe 1 mL, 26 G needle). Two weeks after priming, the secondary immunization was effected and subsequent boosters were performed at one-week intervals to effect a total of 4 or 5 immunizations. For the final immunization, GRP78 (50 [tg) was suspended in 100 [d of PBS and injected into the tail vein; three days later, cell fusion was practiced. 1-3. Constructing hybridomas [0169] Cell fusion was performed in the following manner. The spleen was asceptically - 55 extracted from each mouse and disintegrated in medium 1 (RPMI1640 + PS) to form a suspension of single cells. The suspension was passed through a 70-[Lm nylon mesh (Falcon) to remove the fat tissue and the like before counting the number of cells. The obtained B cells were mixed with mouse myeloma cells (P3U1 cells) such that the ratio of their cell counts was approximately 2:1; 1 mL of 50% PEG (Roche, cat #: 783 641) was then added to effect cell fusion. The fused cells were suspended in medium 2 [RPMI1640 + PS, 10% FCS, HAT (Sigma, H0262), 5% BM condimed Hi (Roche: #1088947)] and distributed in a suitable number (10) of 96-well plates in a volume of 200 [tL/well and cultured at 37"C. One week later, the culture supernatant was used to screen the hybridomas. Example 2: Screening for Anti-GRP78 Antibodies that Recognize GRP78 Localized on the Cell Membrane 2-1. ELISA screening for GRP78 binding antibodies (primary screening) [0170] To acquire anti-GRP78 antibodies localized on the cell surface, screening for GRP78 binding antibodies was first performed with ELISA. [0171] An ELISA plate (NUNC) coated with 1 [tg/ml of the GRP78 protein purified from E. coli was reacted with the culture supernatant of hybridomas and incubated for 1 hour. Thereafter, reaction with alkali phosphatase (AP) labeled anti-mouse IgG (ZYMED: #62-6622) was performed for 1 hour, followed by adding 1 mg/ml of a substrate (SIGMA: S0942-50TAB) to develop color. Using a plate reader (BioRad), OD 4 05 measurement was effected to select ELISA-positive wells. 2-2. FACS screening for antibodies against GRP78 localized on the cell surface (secondary screening) [0172] The culture supernatant in the wells that were found positive in the primary screening was subjected to FACS analysis for the reactivity with a prostate cancer cell line (DU145). [0173] DU145 (obtained from ATCC) was serially cultured in EMEM (invitrogen) containing 10% FCS, 1 mM sodium pyruvate, and 0.1 mM NEAA. DU145 was stripped off with 1 mM EDTA/PBS, reacted with the culture supernatant of hybridomas, and incubated at - 56 - 4"C for 1 hour. Thereafter, an FITC-labeled anti-mouse IgG antibody (BECKMAN COULTER: PN IM0819) was added and incubated at 4'C for 30 minutes. Thereafter, the activities of the respective culture supernatants of hybridomas for binding to the cell surface of DU145 were analyzed by FACS (Becton Dickinson). 2-3. Limiting dilution [0174] The wells that were found to have any slight activity for binding to DU145 cells in FACS analysis were selected and subjected to limiting dilution (LD) to make monoclones of the hybridomas in each well. Specifically, the number of cells in each well was counted and they were seeded in a 96-well plate to give a density of 3 cells/well. Culture was performed for about 10 days and the culture supernatant in the wells where colonies appeared was subjected to ELISA again and screened for antibody-producing monoclones, with the binding activity used as an index. This series of operations gave 6 clones of GRP78-binding antibody (GA-19 antibody, GA-20 antibody, GA-21 antibody, GA-23 antibody, GA-28 antibody, and GA-31 antibody). 2-4. Determination of subtypes [0175] The antibodies were subtyped using IsoStrip (Roche #1 493 027). For subtyping, the culture supernatant of hybridomas was used after 10-fold dilution with PBS(-). [0176] The respective purified antibodies were subtyped as shown below. Table 1 Antibody Subtype GA-19 G1 GA-20 M GA-21 G3 GA-23 G2a GA-28 G1 GA-31 G1 2-5. Purification of antibodies -57- [0177] From 50 mL of the obtained culture supernatant of hybridomas in monoclones, the respective antibodies were purified; for GA-19 antibody, GA-23 antibody, GA-28 antibody, and GA-31 antibody, the supernatant was loaded on a Hi Trap Protein G HP 1 mL column (Amersham Biosciences #17-0404-01), and for GA-20 antibody and GA-21 antibody which were IgM and IgG3, the supernatant was loaded on an open column packed with 1 ml of Protein L-agarose (SIGMA). The supernatant of hybridomas was adsorbed at a flow rate 1 mL/min and, after washing with 20 mL of 20 mM phosphate buffer (pH 7.0), it was eluted with 3.5 mL of 0.1 M glycine-HCl (pH 2.7). The eluted fractions were recovered in 0.5-ml aliquots into Eppendorf tubes to which 1 M Tris-HCl (pH 9.0) was preliminarily added in 50-puL portions. After OD28Onm measurement, the antibody containing fractions were collected and mixed with PBS(-) to make a total of 2.5 mL, followed by buffer replacement with PBS(-) on a PD-10 column (Amersham Biosciences #17-0851-01). Each of the purified antibodies was passed through a 0.22-um filter (MILLIPORE #SLGV033RS) for making a detailed study of its properties in the following example. Example 3: Analysis of GRP78 Antibodies 3-1. Western blot analysis [0178] To confirm that the obtained antibodies would specifically bind to GRP78 (GST-GRP78) purified from E. coli and to GRP78 expressed in DU145 cells, western blot was performed. [0179] In lane 1, samples of DU145 cells (1 X 106 cells) lysed in 300 ul of a lysis buffer (0.5% NP40, 10 mM tris-HCl, 150 mM NaCl, 5 mM EDTA, pH 7.5) were applied, and in lane 2, GST fused GRP78 protein (0.1 ug) purified from E. coli was applied, and blotting was effected on a PVDF membrane in accordance with the usual procedure. The blots were reacted with the respective antibodies (2 pg/ml), then with a secondary antibody (HRP-labeled anti-mouse IgG), followed by protein detection with an ECL western blot detection reagent (GE Healthcare). As a result, it was confirmed that all the antibodies obtained specifically recognized not only the GST-fused GRP78 protein expressed in E. coli but also the GRP78 protein capable of exogenous expression in cells (FIG. 1). - 58 - 3-2. FACS analysis 3-2-1. Activity for binding to the cell surface of prostate cancer cell line (DU145) [0180] The respective purified anti-GRP78 antibodies were analyzed by FACS to see whether they would stain the cell surface of cancer cells. [0181] DU145 cells stripped with 1 mM of EDTA were incubated with each of the antibodies (10 ug/ml) in an FACS buffer at 4 C for 1 hour. Thereafter, an FITC-labeled anti-mouse IgG antibody (BECKMAN COULTER: PN IM0819) was added and the cells were incubated at 4 *C for 30 minutes. Thereafter, the activity for binding to GRP78 on the surface of DU145 cells was analyzed by FACS (Becton Dickinson). [0182] As a result, two of the obtained six clones of anti-GRP78 antiby (GA-20 antibody and GA-21 antibody) stained DU145 cells (FIG. 2). [0183] It was thus confirmed that GA-20 antibody and GA-21 antibody recognized extracellular epitopes of the GRP78 molecule capable of expression in cancer cells. [0184] The nucleotide sequence of the heavy-chain variable region of the GA-20 antibody is depicted in SEQ ID NO: 4, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 5, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID NO: 6, and the amino acid sequence of that light-chain variable region is depicted in SEQ ID NO: 7. The amino acid sequence of CDR1 in the heavy-chain variable region of the GA-20 antibody is depicted in SEQ ID NO: 8, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 9, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 10, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 11, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 12, and the amino acid sequence of CDR3 is depicted in SEQ ID NO: 13. [0185] The nucleotide sequence of the heavy-chain variable region of the GA-21 antibody is depicted in SEQ ID NO: 14, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 15, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID NO: 16, and the amino acid sequence of the light-chain variable region is depicted in SEQ ID NO: 17. The amino acid sequence of CDR1 in the heavy-chain variable - 59 region of the GA-21 antibody is depicted in SEQ ID NO: 18, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 19, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 20, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 21, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 22, and the amino acid sequence of CDR3 is depicted in SEQ ID NO: 23. 3-2-2. Evaluation of FACS binding activity for other cancer types [0186] In the next place, the GA-20 antibody capable of recognizing GRP78 localized on the cell surface was evaluated for its FACS reactivity with other cancer types. Ovary cancer cell lines (ES-2 and SKOV3), a breast cancer cell line (MCF7), a colon cancer cell line (LoVo), and prostate cancer cell lines (DU145, Lncap, 22Rv1 and PC3) were purchased from ATCC and cultured under the ATCC recommended culture conditions. These cells were stained with the GA-20 antibody (10 ug/ml) and subjected to FACS analysis in the above-described manner. As a result, it was confirmed that the GA-20 antibody stained not only DU145 but also other types of cancer cell such as LoVo, LNcap and 22Rv1 (FIG. 3). 3-2-3. Evaluation of FACS binding activity for non-cancer cells [0187] In the next place, the activity for FACS binding to cell lines other than cancer cells was analyzed. Monkey kidney cells (COS7), human normal fibroblast cells (MRC5), mouse pro-B cells (Ba/F3), mouse fibroblast cells (NIH3T3), and hamster ovary cells (DG44) were purchased from ATCC and cultured under the ATCC recommended culture conditions. These cells were stained with the GA-20 antibody (10 ug/ml) and subjected to FACS analysis. At the same time, the cells were lysed with an NP40 lysis buffer and subjected to western blot with the GA-20 antibody (2 pg/ml). As a result, those cells, although they all expressed GRP78 (FIG. 4B), were not stained with FACS (FIG. 4A). From this result, it was speculated that only certain types of cells such as cancer cells would express GRP78 and allow it be localized on the cell surface. 3-3. Analysis for internalizing activity 3-3-1. Analysis by FACS [0188] To determine whether the anti-GRP78 recognizing antibodies that stained the cell -60surface (GA-20 antibody and GA-21 antibody) had an internalizing activity, the following experiment was conducted. [0189] DU145 cells were stripped with 1 mM of EDTA and divided into two groups; the cells in one group were reacted with each antibody (10 ug/ml) in an FACS buffer (PBS containing 2% FCS and 0.05% NaN 3 ) at 0 C for 2 hours, and the cells in the other group were incubated with each antibody (10 pg/ml) in a culture medium (RPMI1640 containing 10% FCS) at 37 C for 2 hours. Thereafter, FACS analysis was performed with FITC-labeled mouse IgG to detect any antibodies that remained on the cell surface. [0190] As a result, it was confirmed that either antibody had disappeared from the cell surface upon 2-hr reaction at 37 'C (FIG. 5). 3-3-2. Analysis by immunocytostaining [0191] To confirm that the phenomenon described above was not extracelluar release of the antibodies, the following experiment was conducted. To DU145 cells being cultured in a dish, the GA-20 antibody or the GA-31 antibody (negative control, no binding activity to the cell surface) was added in 20 pg/ml and cultured at 37 *C for 3 hours. After washing with PBS, the cells were washed twice with a glycine buffer (0.1 M glycine, pH 2.7) to remove the antibodies binding to the cell surface. Thereafter, 4% paraformaldehyde was allowed to act on the cells at room temperature for 10 minutes to immobilize them and, subsequently, 0.1% Triton X100 was allowed to act at room temperature for 5 minutes. The cells were stained with FITC-labeled mouse IgG and the antibodies incorporated into the cells were examined with a fluorescence microscope (KEYENCE). [0192] As a result, the GA-20 antibody having an activity for binding to the cell surface was detected within the cells but the GA-31 antibody not binding to the cell surface was not detected witin the cells (FIG. 6). Thus, it was confirmed that the GA-20 antibody bound to the cell surface had been incorporated into the cells as the result of 3-hr culture at 37'C. Example 4: Epitope Analysis 4-1. Preparation of GST-fused proteins for epitope mapping 4-1-1. Construction of vectors for expressing GST-fused proteins for epitope mapping - 61 - [0193] As the first step in the process of identifying the epitopes of various antibodies, E coli expression vector encoding each part of GRP78 fused to glutathione S-transferase (GST) protein was constructed as bellows. 4-1-1-1. Constructing a GST-GRP78-N(19-350) expressing vector [0194] With pGEX-GRP78-full used as a template, and with GRP-GST-1 (SEQ ID NO: 28) as a sense primer and GRP-GST-3 (SEQ ID NO: 30) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94'C for 30 sec, 64'C for 30 sec, and 72*C for 120 sec, to give a cDNA fragment coding for GRP78 (amino acids 19-350) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0195] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78-N (19-350)). 4-1-1-2. Constructing a GST-GRP78-C (289-654) expressing vector [0196] With pGEX-GRP78-full used as a template, and with GRP-GST-4 (SEQ ID NO: 31) as a sense primer and GRP-GST-2 (SEQ ID NO: 29) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94 *C for 30 sec, 64 'C for 30 sec, and 72 *C for 120 sec, to give a cDNA fragment coding for GRP78 (amino acids 289-654) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0197] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78-C (289-654)). 4-1-1-3. Constructing a GST-GRP78-C (289-350) expressing vector [0198] With pGEX-GRP78-full used as a template, and with GRP-GST-4 (SEQ ID NO: 31) as a sense primer and GRP-GST-3 (SEQ ID NO: 30) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94 *C for 30 sec and 72 *C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 289-350) with BamHI and XhoI cleavage - 62 sequences attached to the 5' and 3' ends, respectively. [0199] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an . coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78-C (289-350)). 4-1-1-4. Constructing a GST-GRP78-C (289-445) expressing vector [0200] With pGEX-GRP78-full used as a template, and with GRP-GST-4 (SEQ ID NO: 31) as a sense primer and GRP-GST-5 (SEQ ID NO: 32) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94 C for 30 sec and 72 C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 289-445) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0201] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78-C (289-445)). 4-1-1-5. Constructing a GST-GRP78-C(289-538) expressing vector expressing vector [0202] With pGEX-GRP78-full used as a template, and with GRP-GST-4 (SEQ ID NO: 31) as a sense primer and GRP-GST-6 (SEQ ID NO: 33) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94 C for 30 sec and 72 *C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 289-538) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0203] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an F. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78-C (289-538)). 4-1-1-6. Constructing a GST-GRP78 (345-385) expressing vector [0204] With pGEX-GRP78-full used as a template, and with GRP-GST-7 (SEQ ID NO: 34) as a sense primer and GRP-GST-8 (SEQ ID NO: 35) as an antisense primer, PCR was - 63 performed in 25 cycles, each consisting of 94*C for 30 sec, 64*C for 30 sec, and 72 0 C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 345-385) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0205] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78 (345-385)). 4-1-1-7. Constructing a GST-GRP78 (376-415) expressing vector [0206] With pGEX-GRP78-full used as a template, and with GRP-GST-9 (SEQ ID NO: 36) as a sense primer and GRP-GST-10 (SEQ ID NO: 37) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94 0 C for 30 sec, 64 0 C for 30 sec, and 72*C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 376-415) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0207] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78 (376-415)). 4-1-1-8. Constructing a GST-GRP78 (406-445) expressing vector [0208] With pGEX-GRP78-full used as a template, and with GRP-GST-11 (SEQ ID NO: 38) as a sense primer and GRP-GST-5 (SEQ ID NO: 32) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94*C for 30 sec, 64'C for 30 sec, and 72'C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 406-445) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0209] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78 (406-445)). 4-1-1-9. Constructing a GST-GRP78 (345-445) expressing vector - 64 - [0210] With pGEX-GRP78-full used as a template, and with GRP-GST-7 (SEQ ID NO: 34) as a sense primer and GRP-GST-5 (SEQ ID NO: 32) as an antisense primer, PCR was performed in 25 cycles, each consisting of 94*C for 30 sec, 64'C for 30 sec, and 72C for 30 sec, to give a cDNA fragment coding for GRP78 (amino acids 345-445) with BamHI and XhoI cleavage sequences attached to the 5' and 3' ends, respectively. [0211] The fragment was cleaved with BamHI and XhoI and inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) that had been similarly cleaved with BamHI and XhoI to construct a GRP78-GST fusion protein expressing vector (pGEX-GRP78 (345-445)). [0212] The sequences of the primers used in constructing the various expression vectors are depicted below. GRP-GST-1: aaaggatccg aggaggagga caagaaggag gacgtggg (SEQ ID NO: 28) GRP-GST-2: tttctcgagc tacaactcat ctttttctgc tgtatcctc (SEQ ID NO: 29) GRP-GST-3: tttctcgagc taatcagaat cttccaacac tttctggacg ggc (SEQ ID NO: 30) GRP-GST-4: aaaggatccc ggcgcgaggt agaaaaggcc aaac (SEQ ID NO: 31) GRP-GST-5: ttctcgagct aggtaggcac cactgtgttc cttgg (SEQ ID NO: 32) GRP-GST-6: ttctcgagct agatttcttc aggtgtcagg cgatt (SEQ ID NO: 33) GRP-GST-7: tttggatccg tgttggaaga ttctgatttg aaga (SEQ ID NO: 34) GRP-GST-8: ttctcgagct aggatggttc cttgccattg aagaa (SEQ ID NO: 35) GRP-GST-9: aaaggatcca aagagttctt caatggcaag ga (SEQ ID NO: 36) GRP-GST-10: ttctcgagct ataccaggtc acctgtatct tgatc (SEQ ID NO: 37) GRP-GST-1 1: aaaggatcct ctggtgatca agatacaggt gac (SEQ ID NO: 38) 4-1-2. Inducing the expression of the respective GST fused GRP78 proteins [0213] Using the thus constructed E. coli expression vectors, E. coli strain BL21 was transformed. The E. coli transformants were cultured in an LB medium (1 ml for each culture) and at the logarithmic growth stage, IPTG (final concentration: 1 mM) was added to induce protein expression. Four or five hours later, the E. coli cells were recovered and lysed in an SDS sample buffer (0.5 ml) to form a lysate; 5 pl of the lysate was taken and in - 65 accordance with the usual practice, it was subjected to SDS-PAGE and subsequently blotted on a PVDF membrane for use in western blotting. 4-2. Epitope mapping of the respective antibodies [0214] The thus prepared GST fused proteins representing the various regions of the GRP78 protein were subjected to western blotting to see which regions of the GRP78 protein would be recognized by the respective GRP78 antibodies obtained. [0215] The result of the first western blotting (FIG. 7) shows that GA-19 antibody, which did not stain cell surface by FACS, recognized half (19-350) of the GRP78 protein toward the N terminus, whereas GA-23 antibody, GA-28 antibody, and GA-31 antibody, which also showed inabllity to bind to cell surface, recognized the 538-654 region toward the C terminus. [0216] In contrast, GA-20 antibody and GA-21 antibody that are capable of binding to cell surface by FACS were found, in view of their stain patterns in western blotting, to recognize a 350-445 region spanning nearly 100 amino acids. [0217] In the next place, GST fused proteins were constructed by dividing the 350-445 region into three areas and subjected to western blotting in the same manner as described above so as to identify the areas to which the GA-20 andtibody and the GA-21 antibody would bind. As a result, the epitopes of these antibodies were found to be the 40 amino acids with amino acid numbers 376-415 in the GRP78 protein (FIG. 8). Example 5: Constructing a Cell Death Inducer Using Extracellular Region Recognizing Anti-GRP78 Antibbody (GA-20 Antibody) 5-1. Cloning the variable regions of GA-20 antibody and analyzing their amino acid sequences [0218] Total RNA was purified from ca. 5 x 106 hybridomas using Trizol (#15596-018, Life technologies). From 1 ug of the total RNA obtained, full-length cDNA was synthesized using SMART RACE cDNA Amplification Kit (CLONTECH #PT3269-1) in accordance with the accompanying manual. With the obtained cDNA used as a template and using Advantage 2 PCR Enzyme System (CLONTECH #PT3281-1), PCR was performed under the following conditions to amplify the genes coding for the heavy-chain variable region (VH) -66and the light-chain variable region (VL) of the GA-20 antibody. Primer for cloning the light-chain variable region Universal primer mix (UPM) to k(VL-k) UPM: Attached to the Kit VL-k: gct cac tgg atg gtg gga aga tg (SEQ ID NO: 39) Primer for cloning the heavy-chain variable region UPM to VH-M UPM: Attached to the Kit VH-M: cca cca gat tct tat cag aca gg (SEQ ID NO: 40) 94 'C x 5 sec and 72 *C x 2 min, 5 cycles; 94 *C x 5 sec, 70 'C x 10 sec, and 72 *C x 2 min, 5 cycles; 94 *C x 5 sec, 68 C x 10 sec, and 72 *C x 2 min, 27 cycles. [0219] The gene fragments amplified by the above procedure were TA-cloned in pCRII TOPO (Invitrogen TOPO TA-cloning kit, #45-0640) and, thereafter, the respective inserts were checked for their nucleotide sequences by an AB13730 sequencer. 5-2. Constructing toxin-labeled GA-20_single-chain Fv antibody (GA20_PE40) 5-2-1. Constructing a GA20_PE40 expressing vector 5-2-1-1. Constructing pET22bHisPE40 [0220] Based on the GA-20 antibody specifically recognizing GRP78 localized on the cell surface, an attempt was made to construct cell death inducing antibodies labeled with an immunotoxin (PE40). [0221] First, an expression vector was constructed that coded for a toxin labeled antibody (GA20_PE40) having a toxin (PE40) attached to a single-chain Fv (scFv) derived from GA-20 antibody. The immunotoxin PE40 gene was amplified by PCR under the following conditions with plasmid DNA (pJH8) purchased from ATCC and used as a template. [0222] Using a sense primer (PE-1) having an EcoRI recognition sequence and a linker sequence (18 amino acids) attached in that order to the 5' end and an antisense primer (PE-2) having a NotI recognition sequence, a termination codon, an ER transition signal sequence -67- (KDEL) and a FLAG tag sequence attached in that order to the 5' end, PCR amplification was performed in a KOD-Plus buffer (2 mM dNTPs, 25 mM MgSO4, and KOD-Plus) (Takara) under the following conditions: 98 *C x 10 sec, 72 'C x 5 sec, 68 'C x 4 min, 5 cycles; 98 C x 10 sec, 70 *C x 5 sec, 68 'C x 4 min, 5 cycles; 98 C x 10 sec, 68 *C x 4 sec, 25 cycles. [0223] The primer sequences are as depicted below. PE-1: taagaattcg gtggcgcgcc ggagttcccg aaaccgtcca ccccgccggg ttcttctggt ttagagggcg gcagcctggc cgcgctg (SEQ ID NO: 41) PE-2: acttagcggc cgctcactac agttcgtctt tcttatcgtc gtcatccttg tagtccggcg gtttgccggg ctggc (SEQ ID NO: 42) [0224] The product amplified by PCR was inserted into pGEM-T easy using pGEM-T Easy Vector System I (Promega). The sequence of the product was confirmed by an AB13730 sequencer. [0225] In the next place, a His tag sequence, a HindIl recognition sequence, an EcoRI recognition sequence, and a NotI recognition sequence were insereted downstream of a PelB signal sequence in an E. coli expression vector pET22b vector (product of Novagen) to construct pET22b_His. [0226] Then, the PE40 gene fragment cloned in pGEM-T easy was digested with EcoRI and NotI, and was sliced out of an agarose gel, followed by insertion of the resulting gene fragment between EcoRI and NotI in pET22bHis to construct pET22bHisPE40. 5-2-1-2. Constructing pET22bHisGA20scFv-PE40 [0227] A gene fragment coding for a single-chain Fv derived from the GA-20 antibody, namely, the heavy-chain variable region of the GA-20 antibody is ligated to its light-chain variable region by a linker sequence composed of 15 amino acids ((GlyGlyGlyGlySer) 3 ) (SEQ ID NO: 43), was amplified by performing PCR under the following conditions. [0228] First, with the heavy-chain variable region of the GA-20 antibody being used as a template after it was TA-cloned in pCRII-TOPO, and using the sense primer GA20-1 (SEQ - 68 - ID NO: 44) and the antisense primer GA20-2 (SEQ ID NO: 45) for the heavy-chain variable region while using the sense primer GA20-3 (SEQ ID NO: 46) and the antisense primer GA20-4 (SEQ ID NO: 47) for the light-chain variable region, PCR amplification was performed by carrying out a reaction with pyrobest DNA polymerase (TAKARA #R005) at 94 C for one minute, followed by 25 cycles of 94 'C for 30 min and 72 *C for 30 min. [0229] In the next place, the thus obtained PCR products of the heavy- and light-chain variable regions were purified on an S-300 HR column (Amersham Biosciences #27-5130 01); the respective products were mixed in 1 [d portions in the same tube and after performing a reaction with pyrobest DNA polymerase at 94 *C for one minute, an annealing reaction was carried out in 5 cycles of 94 *C for 30 min and 72 C for 30 min. [0230] After annealing, 1-[tL of the reaction solution was subjected to PCR amplification using the primers GA20-1 (SEQ ID NO: 44) and GA20-4 (SEQ ID NO: 47) under the following conditions by first performing a reaction with pyrobest DNA polymerase at 94 'C for one minute, then repeating 25 cycles of 94 *C for 30 min and 72 C for 1 min. [0231] The amplified fragment was purified on an S-400 HR column (Amersham Biosciences #27-5140-01), cleaved with EcoRI-HindIII, and sliced out of an agarose gel. The sliced fragment was inserted between HindlIl and EcoR in the pET22bHisPE40 constructed in 5-2-1-1; following confirmation of its nucleotide sequence, pET22b_His_GA20scFv-PE40 was constructed. [0232] The sequences of the primers used in PCR amplification are depicted below. GA20-1: aaaagcttga ggtccagctg caacagtctg g (SEQ ID NO: 44) GA20-2: cccgaaccac caccacccga accaccacca cctgaggaga cggtgactga ggttcc (SEQ ID NO: 45) GA20-3: tggttcgggt ggtggtggtt cgggtggtgg cggatcggac attgtgatgt cacagtctcc atcct (SEQ ID NO: 46) GA20-4: ttgaattctt tgatttccag cttggtgcct c (SEQ ID NO: 47) [0233] The nucleotide sequence of the obtained GA20_PE40 is depicted in SEQ ID NO: 24 and the amino acid sequence prescribed by that nucleotide sequence is depicted in SEQ ID - 69 - NO: 25. 5-2-2. Purifying toxin-labeled GA-20_single-chain Fv antibody (GA20_PE40) [0234] E. coli strain BL21 transformed with pET22b_HisGA20scFv-PE40 was seeded in an LB agar plate containing 50 ug/ml of ampicilin. Grown single colonies were picked up and cultured in an LB medium (3 ml) containing 50 ug/ml of carbenicillin (COSMO BIO). After 4-hr culture, the grown cells were extended in 200 ml of an LB medium containing carbenicillin (50 ug/ml) and cultured continuously. When the growth reached a logarithmic stage, the culture medium was replaced by a new LB medium (200 ml, supplemented with carbenicillin) and IPTG (final 1 mM) was added to induce protein expression. After 5-hr culture, the cells were recovered by centrifugation. [0235] In the presence of a protease inhibitor, Complete EDTA Free (Roche), the cells were suspended in 20 ml of B-BER (PIERCE) for lysis; subsequently, the insoluble proteins were removed by centrifugation to prepare a cell lysate. This sample of lysed cells was applied to a HisTrap column (HiTrap chelating HP 1 ml; Amersham Pharmacia) and GA20PE40 was purified in accordance with the accompanying protocol. To be more specific, the proteins adsorbed on the column were washed with a binding buffer (20 mM sodium phosphate, 0.5 M NaCl, 10 mM imidazole, pH 7.4) and eluted with an elution buffer (20 mM sodium phosphate, 0.5 M NaCl, 500 mM imidazole, pH 7.4) to give a total of 7 fractions each weighing 500 1 . [0236] To determine which of the eluted fractions cotained the desired toxin-labeled GA20 antibody, they were subjected to ELISA assay, with the activity for binding to GRP78 being used as an index, for investigating the binding activities of the eluted fractions. ELISA was practiced in the following manner. To a plate (NUNC) coated with either GST-GRP78 purified from E coli or a negative control HB-EGF protein (R&D) in an amount of 1 pg/ml, each of the fractions as diluted 40-fold with a diluent buffer (1% BSA, 50 mM Tris, 1 mM MgCl 2 , 150 mM NaCl, 0.05% Tween 20) was added. After reaction at room temperature for 1 hour, the plate was washed three times with TBS-T (TBS-0.05% Tween 20) and an anti-flag antibody (M2 antibody, Sigma) was added in an amount of 1 [tg/ml, followed by - 70 incubation at room temperature for 1 hour. After three additional washings with TBS-T, reaction with an alkali phosphatase-labeled anti-mouse IgG (ZYMED) was conducted for 1 hour and a substrate (Sigma) was added in an amount of 1 mg/ml to develop color. [0237] As a result, binding activity specific for GRP78 was recognized in eluted fractions 2 and 3 (elutes 2 and 3), confirming the enrichment of GA20-PE40 protein in elutes 2 and 3 (FIG. 9, lower panel). [0238] Hence, elutes 2, 3 and 4 were loaded on a PD-10 column (GE Healthcare) for buffer replacement by PBS in accordance with the accompanying instruction manual. The elutes were then passed through a 0.
2 2 -pm filter (Millipore) for sterilization and subjected to a study of cytotoxic activity. 5-3. Analyzing the antitumor activity of the toxin-labeled anti-GRP78 antibody (GA20-PE40) [0239] The GA20-PE40 obtained was analyzed for cell death inducing activity. [0240] Hamseter ovary cell line DG44 and prostate cancer cell lines DU145 and 22Rv1 were seeded in amounts of 90 I/well on 96-well plates (1 X 10 3 /well for DG44, and 6 X 103 /well for DU145 and 22Rv1). On the next day, GA20-PE40 fractions (elutes 2, 3, and 4) and PBS were added to the plates in amounts of 10 R/well, and culture was performed at 37'C. Five days later, viable cell counts were taken with the reagent WST-8 (DOJINDO LABORATORIES) and compared with the data for the control (PBS treated group); the results were numerically represented in graphs. [0241] As it turned out, the cell growth of DG44 was not at all affected by GA20-PE40 (FIG. 10C); on the other hand, elutes 2 and 3 which were found to be active for binding to GRP78 were recognized to have cytotoxic activity in the two prostate cancer cell lines (DU145 and 22Rv1) (FIG. 10A for DU145 and FIG. 10B for 22Rv1). [0242] These results revealed that the antibody against the extracellular epitope (350-445) of GRP78 is useful as an antitumor agent. Example 6: Acquisition of Anti-GRP78 Antibodies by Re-immunization 6-1. Preparation of immunogen GST-GRP78 (376-415) protein - 71 - [0243] From the foregoing analyses described in Example 4, it was speculated that the 3 7 6 th to 4 15 th amino acids in the GRP78 protein would form an extracellular region of GRP78. Hence, the present inventor re-immunized that region in an attempt to establish antibodies having strong enough affinity to recognize the extracellular region of GRP78. [0244] First, E coli (BL21) cells transformed with the GST-GRP78(376-415) expressing vector (pGEX-GRP78 (376-415)) described in Example 4 were cultured in an LB medium (250 ml) and when an OD 610 value of at least was reached, induction of protein expression was effected by means of IPTG (1 mM). After 5-hr culture, the E coli cells were harvested and lysed in 25 ml of B-PER (PIERCE). The E coli lysate was then diluted 10-fold with PBS; to the diluted lysate, PBS-equilibrated glutathione Sepharose 4B (Amersham Pharmacia) was added, followed by incubation at 4 C overnight. Thereafter, the glutathione Sepharose 4B was washed with PBS several times to remove any unadsorbed proteins and the desired protein was eluted with 20 mM glutathione. [0245] The eluted fractions were subjected to SDS-PAGE and stained with CBB to collect the fractions containing the desired protein. The sample was further separated from impure proteins in PBS by gel filtration chromatography (Superdex 200 16/60, GE Healthcare) and only the desired protein was purified to higher purity. The purified protein was used as an immunogen in the following experiment. 6-2. Immunization of GST-GRP78(376-415) protein [0246] The GST-GRP78(376-415) purified in 6-1 was applied by the same technique as used in Example 1 (1-2) to immunize mice in the following groups [(MRL/lpr, male, 4-wk old) (Balb/c, female, 6-wk old): all purchased from charles river, Japan]. Hybridomas were constructed as described in Example 1 (1-3). 6-3. Screening for anti-GRP78 atibodies 6-3-1. Purification of MBP-GRP78(376-415) protein [0247] A fusion protein (MBP-GRP78 (376-415)) was prepared from GRP78 ( 3 7 6 th to 415t amino acids) and a maltose binding protein (MBP) in the following manner. [0248] The pGEX-GRP78(376-415) constructed in Example 4 was cleaved with - 72 - BamHI-SalI to slice a gene fragment coding for GRP78 (376-415). The fragment was inserted between BamHI and SalI in pMAL-c2X (New England BioLabs) to construct the MBP-GRP78(376-415) expressing vector pMAL-c2X-GRP78 (376-415). [0249] In the next place, E. coli strain BL21 transformed with this vector was cultured in an LB medium (250 ml) and when an OD 610 value of at least 0.5 was reached, IPTG (1 mM) was used to induce protein expression. After 5-hr culture, the E. coli cells were harvested by centrifugation and lysed with 25 ml of B-PER (PIERCE). Then, the E. coli lysate was diluted 5-fold with a column buffer (20 mM Tris, pH 7.5, 200 mM NaCl, 1 mM EDTA); to the diluted lysate, an amylose resin (New England BioLabs) equilibrated with the column buffer was added and incubated at 4 C overnight. Subsequently, the amylose resin was washed with the column buffer several times to remove any unadsorbed proteins, and the desired protein was eluted with an elution buffer (the column buffer containing 10 mM maltose). The eluted fractions were subjected to SDS-PAGE and the fractions containing the desired protein were identified by CBB stain, combined into one fraction, and loaded on a PD10 column to effect buffer replacement with PBS. The thus purified MBP-GRP78(376 415) was used in the following experiment as a sample for ELISA screening for binding antibodies. 6-3-2. ELISA screening for GRP78 binding antibodies (primary screening) [0250] An ELISA plate coated with 1 [tg/ml of the MBP-GRP78 (376-415) purified in 6-3-1 was used to screen for antibodies that would bind to the region consisting of the 3 76 th to 4 15 th amino acids in the GRP78 protein. [0251] The screening method was as described in Example 2 (2-1). The GRP78 binding antibodies obtained by primary screening, with the activity for binding to the MBP-GRP78 (376-415) protein used as an index, were subjected to subsequent secondary screening, as described below. 6-3-3. FACS screening for anti-GRP78 antibodies localized on the cell surface (secondary screening) [0252] The GRP78 binding antibodies obtained by the primary screening were then - 73 subjected to secondary screening, with the activity for binding to prostate cancer cell lines (DU145 and 22Rv1 (ATCC CRL-2505)) being used as an index. The method was as described in Example 2 (2-2). [0253] As a result, four additional antibodies, GC-18 antibody, GC-20 antibody, GD-4 antibody, and GD-17 antibody, were found to be capable of staining the prostate cancer cell lines by FACS. [0254] These antibodies were subjected to limiting dilution by the method described in Example 2 (2-3) to form monoclones. The antibodies were subtyped by the method described in Example 2 (2-4). The subtypes of the respective antibodies are as shown below. Table 2 Antibody Subtype GC-18 G1 GC-20 G1 GD-4 G1 GD-17 G1 [0255] Subsequently, the antibodies were purified by the method described in Example 2 (2-5), and analyzed for their properties in detail as follows. 6-4. Analyzing the additional antibodies obtained by re-immunization 6-4-1. FACS analysis [0256] To confirm that the purified four antibodies would stain cell surfaces of cancer cells, FACS analysis was carried out using a prostate cancer cell line (22Rv1). The cells were stained with each antibody (10 Rg/ml) and subjected to FACS analysis by the method described in Example 3 (3-2-1). [0257] As a result, all those antibodies were confirmed to bind to cell surfaces of 22Rv1 cells (FIG. 11). 6-4-2. Epitope mapping [0258] Since the four antibodies obtained above were established by immunizing the - 74 - GST-GRP78 (376-415) protein, they are antibodies that recognize the partial region between the 3 7 6 th and 4 1 5th amino acids in GRP78. Thus, this region was further divided into four areas, namely, the 376th to 391st amino acids (i.e., amino acids 1-16 in SEQ ID NO: 3), the 384to 3 9 9 th amino acids (i.e., amino acids 9-24 in SEQ ID NO: 3), the 3 9 2 "d to 4 0 7 th amino acids (i.e., amino acids 17-32 in SEQ ID NO: 3), and the 4 0 0 th to 4 15 th amino acids (i.e., amino acids 25-40 in SEQ ID NO: 3), and an analysis was made to see which part of the sequence composed by the 3 7 6 th to 4 15 th amino acids in GRP78 was recognized by each of those four antibodies. 6-4-2-1. Preparing GST fused proteins for epitope mapping 6-4-2-1-1. Constructing vectors for expressing the GST fused proteins for epitope mapping [0259] DNA fragments coding for the 3 7 6 th to 391st amino acids, 3 8 4 th to 3 9 9 th amino acids, 3 9 2 "d to 4 0 7 h amino acids, and the 4 0 0 th to 4 1 5th amino acids, respectively, in the GRP78 protein were constructed in the following manner. [0260] A DNA fragment coding for GRP78(376-391) (i.e., amino acids 1-16 in SEQ ID NO: 3) was constructed by annealing the oligomers GEP1/GEP2 (as depicted in SEQ ID NOS:48 and 49, respectively), a DNA fragment coding for GRP78(384-399) (i.e., amino acids 9-24 in SEQ ID NO: 3) was constructed by annealing the oligomers GEP3/GEP4 (as depicted in SEQ ID NOS:50 and 51, respectively), a DNA fragment coding for GRP78(392 407 (i.e., amino acids 17-32 in SEQ ID NO: 3) was constructed by annealing the oligomers GEP5/GEP6 (as depicted in SEQ ID NOS:52 and 53, respectively), and a DNA fragment coding for GRP78(400-415) (i.e., amino acids 25-40 in SEQ ID NO: 3) was constructed by annealing the oligomers GEP7/GEP8 (as depicted in SEQ ID NOS:54 and 55, respectively). [0261] The sequences of the oligomers used to construct those DNA fragments are shown below. GEP1: gatccaaaga gttcttcaat ggcaaggaac catcccgtgg cataaaccca gatc (SEQ ID NO: 48) GEP2: tcgagatctg ggtttatgcc acgggatggt tccttgccat tgaagaactc tttg (SEQ ID NO: 49) - 75 - GEP3: gatccccatc ccgtggcata aacccagatg aagctgtagc gtatggtgct gctc (SEQ ID NO: 50) GEP4: tcgagagcag caccatacgc tacagcttca tctgggttta tgccacggga tggg (SEQ ID NO: 51) GEP5: gatccgaagc tgtagcgtat ggtgctgctg tccaggctgg tgtgctctct ggtc (SEQ ID NO: 52) GEP6: tcgagaccag agagcacacc agcctggaca gcagcaccat acgctacagc ttcg (SEQ ID NO: 53) GEP7: gatccgtcca ggctggtgtg ctctctggtg atcaagatac aggtgacctg gtac (SEQ ID NO: 54) GEP8: tcgagtacca ggtcacctgt atcttgatca ccagagagca caccagcctg gacg (SEQ ID NO: 55) [0262] Each of the DNA fragments thus constructed was inserted downstream of the GST coding region of an E. coli expression vector (pGEX-6P-1) cleaved with BamHI and XhoI, thereby constructing GRP78-GST fused protein expressing vectors (respectively designated pGEX-GRP78 (376-391), pGEX-GRP78 (384-399), pGEX-GRP78 (392-407), and Pgex-GRP78 (400-415)). 6-4-2-1-2. Inducing the expression of the respective GST fused GRP78 proteins [0263] The thus constructed E. coli expression vectors were used to transform E coli strain BL21 and protein expression was inducted by the method describeD in Example 4 (4-1-2). As a result, it was confirmed that the desired priotein was expressd in E. coli, as shown in FIG. 12B. Hence, this protein was used in epitope mapping as follows. 6-4-2-2. Epitope mapping of the respective antibodies [0264] The thus prepared GST fused proteins were subjected to western blotting to see which regions of the GRP78 protein would be recognized by the respective GRP78 antibodies obtained. [0265] The stain patterns of western blot (FIG. 13) revealed that the GD-17 antibody recognized the region spanning the 3 8 4 th to 391" amino acids in GRP78, the GC-18 and GC-20 antibodies recognized the region spanning the 3 9 2 "d to 4 0 7 th amino acids, and the -76- GD-4 antibody recognized the region of the 4 0 0 th to 4 1 5 th amino acids. It was also found that the GA-20 antibody obtained at the earlier stage recognized the same region as the GD-4 antibody (FIG. 13). 6-4-3. Cloning the variable regions and determining their amino acid sequences [0266] The variable regions of the additionally obtained antibodies (GC-18 antibody, GC-20 antibody, GD-4 antibody, and GD-17 antibody) were cloned and analyzed for their animo acid sequences by the methods described in Example 5 (5-1). It should, however, be noted that since all those antibodies were IgG 1 , the variable regions of their heavy chains were cloned using the following VH-G1 primer (SEQ ID NO: 56): [0267] VH-G1: cca cca gat tct tat cag aca gg (SEQ ID NO: 56) [0268] The amplified light-chain and heavy-chain gene fragments were TA-cloned in pCRII-TOPO (Invitrogen TOPO TA-cloning kit, #45-0640) and, thereafter, the respective inserts were checked for their nucleotide sequences. [0269] The nucleotide sequence of the heavy-chain variable region of the GC-18 antibody binding to the region spanning the 3 9 2 "d to 4 0 7 th amino acids in GRP78 is depicted in SEQ ID NO: 57, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 58, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID NO: 59, and the amino acid sequence of that light-chain variable region is depicted in SEQ ID NO: 60. The amino acid sequence of CDR1 in the heavy-chain variable region of the GC-18 antibody is depicted in SEQ ID NO: 61, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 62, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 63, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 64, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 65, and the amino acid sequence of CDR3 is depicted in SEQ ID NO: 66. [0270] The nucleotide sequence of the heavy-chain variable region of the GC-20 antibody binding to the region spanning the 3 9 2 nd to 4 0 7 th amino acids in GRP78 is depicted in SEQ ID NO: 67, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 68, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID - 77 - NO: 69, and the amino acid sequence of that light-chain variable region is depicted in SEQ ID NO: 70. The amino acid sequence of CDR1 in the heavy-chain variable region of the GC-20 antibody is depicted in SEQ ID NO: 71, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 72, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 73, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 74, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 75, and the amino acid sequence of CDR3 is depicted in SEQ ID NO: 76. [0271] The nucleotide sequence of the heavy-chain variable region of the GD-4 antibody binding to the region of the 4 0 0 'h to 4 1 5 th amino acids in GRP78 is depicted in SEQ ID NO: 77, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 78, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID NO: 79, and the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 80. The amino acid sequence of CDR1 in the heavy-chain variable region of the GD-4 antibody is depicted in SEQ ID NO: 81, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 82, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 83, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 84, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 85, and the amino acid sequence of CDR3 is depicted in SEQ ID NO: 86. [0272] The nucleotide sequence of the heavy-chain variable region of the GD-17 antibody binding to the region spanning the 3 8 4 th to 391st amino acids in GRP78 is depicted in SEQ ID NO: 87, the amino acid sequence of that heavy-chain variable region is depicted in SEQ ID NO: 88, the nucleotide sequence of the light-chain variable region is depicted in SEQ ID NO: 89, and the amino acid sequence of that light-chain variable region is depicted in SEQ ID NO: 90. The amino acid sequence CDR1 in the heavy-chain variable region of the GD-17 antibody is depicted in SEQ ID NO: 91, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 92, the amino acid sequence of CDR3 is depicted in SEQ ID NO: 93, the amino acid sequence of CDR1 in the light-chain variable region is depicted in SEQ ID NO: 94, the amino acid sequence of CDR2 is depicted in SEQ ID NO: 95, and the amino acid sequence of - 78 - CDR3 is depicted in SEQ ID NO: 96. Example 7: Analyzing the Drug Efficacy of Toxin-Labeled GD17 Single-Chain Antibody (GA17_scFv-PE40) 7-1. Constructing GD17_scFv-PE40 expression vector [0273] A gene fragment coding for a single-chain Fv derived from the GD-17 antibody, namely, the heavy-chain variable region of the GD-17 antibody as ligated to its light-chain variable region by a linker sequence composed of 15 amino acids ((GlyGlyGlyGlySer) 3 ) (SEQ ID NO: 43), was amplified by performing PCR under the following conditions. [0274] First, with the heavy-chain variable region of the GD-17 antibody being used as a template after it was TA-cloned in pCRII-TOPO, and using the sense primer GD17-1 (SEQ ID NO: 97) and the antisense primer GD17-2 (SEQ ID NO: 98) for the heavy-chain variable region while using the sense primer GD17-3 (SEQ ID NO: 99) and the antisense primer GD17-4 (SEQ ID NO: 100) for the light-chain variable region, PCR amplification was performed by carrying out a reaction with pyrobest DNA polymerase (TAKARA#R005) at 94 C for one minute, followed by 25 cycles of 94'C x 30 min and 72'C x 30 min. [0275] In the next place, the thus obtained PCR products of the heavy- and light-chain variable regions were purified on an S-300 HR column (Amersham Biosciences #27-5130 01); the respective products were mixed in 1 [1 portions in the same tube and after performing a reaction with pyrobest DNA polymerase at 94 C for one minute, an annealing reaction was carried out in 5 cycles of 94*C x 30 min and 72"C x 30 min. [0276] After annealing, 1- L of the reaction solution was subjected to PCR amplification using the primers GD17-1 (SEQ ID NO: 97) and GD17-4 (SEQ ID NO: 100) under the following conditions by first performing a reaction with pyrobest DNA polymerase at 94'C for one minute, then repeating 25 cycles of 94*C x 30 min and 72'C x 1 min. [0277] The amplified fragmetn was purified on an S-400 HR column (Amersham Biosciences #27-5140-01), cleaved with EcoRI-HindIII, and sliced out of an agarose gel. The sliced fragment was inserted between HindlIl and EcoR in the pET22bHisPE40 constructed in Example 5 (5-2-1-1); following confirmation of its nucleotide sequence, - 79 pET22bHisGD17scFv-PE40 was constructed. [0278] The sequences of the primers used in PCR amplification are shown below. GD17-1: aaaagcttca ggttcagctc cagcagtctg g (SEQ ID NO: 97) GD17-2: cccgaaccac caccacccga accaccacca cctgaggaga ctgtgagagt ggtgcct (SEQ ID NO: 98) GD17-3: tggttcgggt ggtggtggtt cgggtggtgg cggatcggat gttgtgatga cccaaactcc ac (SEQ ID NO: 99) GD17-4: ttgaattctt tcagctccag cttggtccc (SEQ ID NO: 100) [02791 The nucleotide sequence of the obtained GD17scFv_PE40 is depicted in SEQ ID NO: 101 and the amino acid sequence prescribed by that nucleotide sequence is depicted in SEQ ID NO: 102. 7-2. Mass purification of toxin-labeled single-chain antibody [0280] E. coli strain BL21 transformed with pET22bHisGD17scFv-PE40 was cultured in an LB medium containing carbenicillin (50 [tg/ml). When the growth reached a logarithmic stage, IPTG (final dose, 1 mM) was added and culture was effected at room temperature (24'C) overnight to induce protein expression. The E. coli cells recovered by centrifugation were suspended in a binding buffer (20 mM sodium phosphate, 500 mM NaCl, 20 mM imidazole, pH 7.4), disrupted by sonication, and the lysed fragments were applied to a HisTrap FF cride column (GE Heathcare). Thereafter, the dedired protein was eluted with an elution buffer (20 mM sodium phosphate, 500 mM NaCl, 500 mM imidazole, pH 7.4), diluted about 10-fold with a TBS buffer, then applied to an affinity gel packed with an M2 agarose (Sigma). Using an AKTA Explorer (GE Healthcare), the desired protein was eluted with an M2 elution buffer (0.1 M glycine-HCl, pH. 3.5), immediately followed by buffer replacement with PBS on a PD10 column (GE Healthcare) to prepare a final specimen. [0281] The purified GD17scFv-PE40 was subjected to SDS-PAGE, then CBB stain was performed to confirm that it had been purified to 100% purity (FIG. 14). 7-3. Analyzing the activity for binding to the GRP78 protein [0282] In the next place, the purified GD17scFv-PE40 protein was analylzed for its GRP78 - 80 binding activity. Three specimens were prepared, one stored at 4 'C, another left to stand at 37*C overnight, and the third frozen and thawed; their activities for binding to GRP78 in solid phase were measured by ELISA and compared to each other. [0283] Each specimen was diluted with a diluent buffer (1% BSA, 50 mM Tris, 1 mM MgCl 2 , 150 mM NaCl, 0.05% Tween 20) and the dilution was added to a plate (NUNC) coated with the GST-GRP78 (1 [tg/ml) purified from E coli. After reaction at room temperature for 1 hour, the plate was washed three times with TBS-T (TBS-0.05% Tween 20) and an anti-flag antibody (M2 antibody, Sigma) was added in an amount of 1 Rg/ml, followed by incubation at room temperature for 1 hour. After three additional washings with TBS-T, reaction with an alkali phosphatase-labeled anti-mouse IgG (ZYMED) was conducted for 1 hour and a substrate (Sigma) was added in an amount of 1 mg/ml to develop color. [0284] As it turned out, the GD17scFv-PE40 protein, whether it was stored at 4'C or left to stand at 37 'C overnight or subjected to a freeze-thaw cycle, had a GRP78 protein binding activity on the order of EC 50 =1.3 nM, demonstrating that the purified specimens are comparatively stable proteins (FIG. 15). 7-4. Analysis of in vitro cell death inducing activity [0285] In the next place, the activity of the purified GD17scFv-PE40 for inducing cell death was analyzed on cancer cell lines (22Rv1, LNcap, MCF7, BxPC3, PANC1, and SKOV3) or human derived normal cell lines (HUVEC and MRC5) and mouse derived normal cell lines (CHO, NIH3T3, and BaF3). [0286] Among the cell lines used in the experiment, HUVEC was purchased from CAMBREX and the other cell lines were purchased from ATCC, and they were cultured in accordance with the instruction manuals provided by the suppliers. [0287] The cells of each type were seeded on a 96-well plate and, on the next day, GD17scFv-PE40 was diluted at various concentrations in a 10% FCS containing RPMI1640 medium (Invitrogen) and added to the cells. After 5-day culture, the number of viable cells was counted with WST-8 (nakalai). - 81 - [0288] As it turned out, the sensitivity of GD17scFv-PE40 for the cancer cells varied somewhat but it was confirmed to have strong cell death inducing activities of approximately 2-20 nM in terms of EC 5 o value, or the concentration at which it showed 50% of a maximum activity (FIG. 16A). In particular, the EC 5 o values for MCF7 and 22Rv1 cells were on the order of 2-4 nM, thus confirming the potent cytotoxic activity of GD17scFv-PE40. On the other hand, GD17scFv-PE40 was either totally inactive against the human and mouse normal cells or found to have only low cytotoxic activities when it was added at high concentrations (FIG. 16B). [0289] To confirm that this difference in sensitivity was independent of the fact that the GRP78 protein was expressed in the cancer cells but not in the normal cells, western blotting was performed with the GD-17 antibody. Cell lysates were prepared from the respective types of cells in accordance with the conventional practice and after SDS-PAGE, western blotting was conducted with the GD-17 antibody (2 [tg/ml). The result is shown in FIG. 17, from which it is clear that bands specifically stained by the GD-17 were detected in those cell lines on which GD17scFv-PE40 showed no cytotoxic activity; thus, it was assumed that the cancer cell specific cytotoxic activity of GD17scFv-PE40 was not due to the fact that the GRP78 protein was expressed in the cancer cells and not in the normal cells but due to the difference between the two types of cells in terms of localization of the GRP78 protein. 7-5. Analysis of in vivo antitumor activity [0290] The cells of human prostate cancer cell line 22Rv1 (ATCC CRL-2505) were recovered into a 0.05% trypsin supplemented 0.02% EDTA solution and grafted to nude mice [male, 7-wk old (CAnN.Cg-Foxn1<nu>/CrlCrlj (BALB-nu/nu)): charles river, Japan] under the skin of the abdomen in a cell count of 1 X 107 cells/0.2 mL HBSS (SIGMA Cat. No. H 9269). After confirming tumor implantation, the animals were divided into seven groups (one control group, and six drug administered groups) by tumor volume and body weight on the 16 th day of grafting (day 16). [0291] On the day after the grouping (day 17), as well as at days 21, 23, 26, and 29, the control group was administered with physiological saline whereas the drug administered - 82 groups were administered with GD17scFv-PE40 at a dose of 0.5 mg/kg; both physiological saline and GD17scFv-PE40 were intravenously administered instantaneously in an amount of 10 mL/kg. The tumor volume was measured over time until a final measurement was made two days after the final administration (day 31). [0292] The result is shown in FIG. 18. The percent tumor growth suppression at the final measurement was 47% and an analysis of the tumor volume data by nonparametric Dunnet multiple comparison revealed a significant tumor growth suppressing effect in the groups administered with 0.5 mg/kg of GD17scFv-PE40. This result, showing the in vivo efficacy of GD17scFv-PE40 agains the target GRP78, demonstrated the usefulness of the GRP78 targeting antibodies in cancer treatment. INDUSTRIAL APPLICABILITY [0293] It has been shown hereinabove that the present invention, by providing novel antibodies that have an activity for binding to GRP78 and which can internalize, can offer novel pharmaceutical compositions that can be used to treat various tumors and cancers that have GRP78 exposed on the cell surface. In addition, by using the antibodies having such characteristics, methods of diagnosing various tumors and cancers can be provided. - 83 -

Claims (28)

1. A pharmaceutical composition containing an antibody that binds to a glucose regulated protein 78 (GRP78).
2. The composition according to claim 1 which is an anticancer agent.
3. The composition according to claim 1 or 2, wherein the antibody is a monoclonal antibody.
4. The composition according to any one of claims 1 to 3, wherein the antibody binds to GRP78 localized on cell surfaces.
5. The composition according to any one of claims 1 to 4, wherein the antibody is internalized into cells expressing GRP78.
6. The composition according to any one of claims 1 to 5, wherein the antibody binds to the epitope depicted in SEQ ID NO: 3.
7. The composition according to any one of claims 1 to 6, wherein the antibody is conjugated with a cytotoxic substance.
8. A monoclonal antibody that binds to GRP78.
9. The antibody according to claim 8 which binds to GRP78 expressed on cell surfaces.
10. The antibody according to claim 8 or 9 which is internalized into cells expressing GRP78.
11. The antibody according to any one of claims 8 to 10 which binds to the epitope depicted in SEQ ID NO: 3.
12. The antibody according to any one of claims 8 to 11 which recognizes the same epitope as the one that is recognized by an antibody selected from among the following (a) to (f): (a) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 8 as CDR1, the amino acid sequence depicted in SEQ ID NO: 9 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 10 as CDR3, and the light-chain variable region - 84 - having the amino acid sequence depicted in SEQ ID NO: 11 as CDR1, the amino acid sequence depicted in SEQ ID NO: 12 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 13 as CDR3; (b) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 18 as CDR1, the amino acid sequence depicted in SEQ ID NO: 19 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 20 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 21 as CDR1, the amino acid sequence depicted in SEQ ID NO: 22 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 23 as CDR3; (c) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 61 as CDR1, the amino acid sequence depicted in SEQ ID NO: 62 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 63 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 64 as CDR1, the amino acid sequence depicted in SEQ ID NO: 65 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 66 as CDR3; (d) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 71 as CDR1, the amino acid sequence depicted in SEQ ID NO: 72 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 73 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 74 as CDR1, the amino acid sequence depicted in SEQ ID NO: 75 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 76 as CDR3; (e) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 81 as CDR1, the amino acid sequence depicted in SEQ ID NO: 82 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 83 as CDR3, and the light-chain variable - 85 - region having the amino acid sequence depicted in SEQ ID NO: 84 as CDR1, the amino acid sequence depicted in SEQ ID NO: 85 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 86 as CDR3; and (f) an antibody comprising a heavy-chain variable region and a light-chain variable region, the heavy-chain variable region having the amino acid sequence depicted in SEQ ID NO: 91 as CDR1, the amino acid sequence depicted in SEQ ID NO: 92 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 93 as CDR3, and the light-chain variable region having the amino acid sequence depicted in SEQ ID NO: 94 as CDR1, the amino acid sequence depicted in SEQ ID NO: 95 as CDR2, and the amino acid sequence depicted in SEQ ID NO: 96 as CDR3.
13. The antibody according to any one of claims 8 to 12 which has cytotoxic activity against cells expressing GRP78.
14. The antibody according to claim 13 which is conjugated with a cytotoxic substance.
15. A method of delivering a cytotoxic substance into cells using an anti-GRP78 antibody.
16. A method of suppressing the growth of cells using a cytotoxic substance bound to an anti-GRP78 antibody.
17. The method according to claim 15 or 16, wherein the cells are cancer cells.
18. Use of an anti-GRP78 antibody for delivering a cytotoxic substance into cells.
19. Use of an anti-GRP78 antibody having an internalizing activity for suppressing the growth of cells.
20. The use according to claim 18 or 19, wherein the cells are cancer cells.
21. The use according to any one of claims 18 to 20, wherein the cytotoxic substance is bound to the anti-GRP78 antibody.
22. A process for producing a pharmaceutical composition comprising the following steps: (a) the step of providing GRP78 antibodies; (b) the step of validating whether the antibodies of (a) have an internalizing - 86 - activity; (c) the step of selecting antibodies having an internalizing activity; and (d) the step of binding a cytotoxic substance to the antibodies selected in (c).
23. The process according to claim 22, wherein the pharmaceutical composition is an anticancer agent.
24. A method of diagnosing a cancer using an anti-GRP78 antibody.
25. The method of diagnosing according to claim 24 which uses an anti-GRP antibody conjugated with a labeling substance.
26. The method of diagnosing according to claim 24 or 25 which detects the anti-GRP78 antibody incorporated into cells.
27. An anti-GRP78 antibody conjugated with a labeling substance.
28. A polypeptide comprising the amino acid sequence of SEQ ID NO: 3 or a fragment thereof. - 87 -
AU2008219972A 2007-02-27 2008-02-27 Pharmaceutical composition comprising anti-GRP78 antibody as active ingredient Abandoned AU2008219972A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2007-047534 2007-02-27
JP2007047534 2007-02-27
PCT/JP2008/053898 WO2008105560A1 (en) 2007-02-27 2008-02-27 Pharmaceutical composition comprising anti-grp78 antibody as active ingredient

Publications (1)

Publication Number Publication Date
AU2008219972A1 true AU2008219972A1 (en) 2008-09-04

Family

ID=39721380

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008219972A Abandoned AU2008219972A1 (en) 2007-02-27 2008-02-27 Pharmaceutical composition comprising anti-GRP78 antibody as active ingredient

Country Status (7)

Country Link
US (1) US8192740B2 (en)
EP (1) EP2130552B1 (en)
JP (1) JP5374360B2 (en)
CN (1) CN101951953A (en)
AU (1) AU2008219972A1 (en)
CA (1) CA2679266A1 (en)
WO (1) WO2008105560A1 (en)

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7906102B2 (en) 2001-10-03 2011-03-15 Vanderbilt University Ligands to radiation-induced molecules
US7306925B2 (en) 2001-11-09 2007-12-11 Vanderbilt University Phage antibodies to radiation-inducible neoantigens
US20100135904A1 (en) 2008-11-07 2010-06-03 Research Development Foundation Compositions and methods for the inhibition of cripto / grp78 complex formation and signaling
TWI541021B (en) 2009-03-05 2016-07-11 艾伯維有限公司 Il-17 binding proteins
EP2246364A1 (en) * 2009-04-29 2010-11-03 Pierre Fabre Médicament Anti CXCR4 antibodies for the treatment of HIV
GB2475466A (en) * 2009-05-29 2011-05-25 Weiming Xu scFv antibody fragments against GRP78 used for cancer treatment
US20110059111A1 (en) * 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
JP5493714B2 (en) * 2009-10-30 2014-05-14 国立大学法人信州大学 Clathrin-binding peptide derivative
CA2791652C (en) 2010-03-02 2018-06-12 Kyowa Hakko Kirin Co., Ltd. Modified antibody composition
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
WO2012097313A2 (en) 2011-01-14 2012-07-19 The Regents Of The University Of California Therapeutic antibodies against ror-1 protein and methods for use of same
AR087364A1 (en) * 2011-07-29 2014-03-19 Pf Medicament ANTI-CXCR4 ANTIBODY AND ITS USE FOR CANCERES DETECTION AND DIANOSTIC
WO2013019730A1 (en) * 2011-07-29 2013-02-07 The Washington University Antibodies to tip-1 and grp78
BR112015023212A2 (en) 2013-03-14 2017-11-21 Gill Parkash cancer treatment using antibodies that bind to cell surface grp78
JP6682426B2 (en) * 2013-05-24 2020-04-15 メディミューン,エルエルシー Anti-B7-H5 antibody and use thereof
EP3172222A4 (en) 2014-07-24 2018-03-21 Washington University Compositions targeting radiation-induced molecules and methods of use thereof
MA40921A (en) * 2014-11-05 2017-09-12 Abbvie Stemcentrx Llc ANTI-GENETIC CHEMERICAL ANTI-CLDN RECEPTORS AND METHODS OF USE
AU2015357533B2 (en) * 2014-12-05 2021-10-07 Eureka Therapeutics, Inc. Antibodies targeting B-cell maturation antigen and methods of use
WO2018022479A1 (en) * 2016-07-25 2018-02-01 Biogen Ma Inc. Anti-hspa5 (grp78) antibodies and uses thereof
JP6202652B1 (en) * 2016-12-19 2017-09-27 株式会社ヘルスケアシステムズ Anti-equol antibody composition and use thereof
WO2018148595A1 (en) 2017-02-10 2018-08-16 Washington University Antibodies to tip1 and methods of use thereof
WO2019074342A1 (en) * 2017-10-13 2019-04-18 주식회사 나이벡 Method for selecting highly efficient stem cell, using protein marker grp78
CN107998398A (en) * 2017-12-26 2018-05-08 广东省中医院 Application of the GRP78 genes in tumor stem cell chemosensitivity medicine is improved
CA3094534A1 (en) * 2018-03-19 2019-09-26 Abeome Corporation High affinity neutralizing monoclonal antibodies to programmed death ligand 1 (pd-l1) and uses thereof
US10858409B2 (en) * 2018-06-13 2020-12-08 Immune Regulation Limited Protein with anti-inflammatory properties
CA3139057A1 (en) * 2019-06-04 2020-12-10 Marcela V. Maus Antibodies and chimeric antigen receptors that target taci
BR112021025029A2 (en) 2019-06-13 2022-01-25 Allogene Therapeutics Inc Anti-talen antibodies and their uses
EP4031576A1 (en) 2019-09-18 2022-07-27 Molecular Templates, Inc. Pd-l1 binding molecules comprising shiga toxin a subunit scaffolds
WO2021055816A1 (en) 2019-09-18 2021-03-25 Molecular Templates, Inc. Pd-l1 binding molecules comprising shiga toxin a subunit scaffolds
CN113621059B (en) * 2020-05-06 2023-06-06 江苏众红生物工程创药研究院有限公司 Beta-form 2 Microglobulin detection kit and clinical application thereof
KR102511416B1 (en) * 2020-09-18 2023-03-17 주식회사 하울바이오 GRP78 specific antibody and use thereof
US20220251166A1 (en) * 2021-01-29 2022-08-11 Chugai Seiyaku Kabushiki Kaisha Molecule specifically acting in a tissue where a cell death is being observed
CN114163526B (en) * 2021-11-12 2022-06-21 深圳市人民医院 Nano antibody targeting glucose regulatory protein 78 and application thereof
CN115975036B (en) * 2022-08-18 2023-06-09 北京诺赛国际医学研究院 Pharmaceutical composition comprising stem cells and use thereof for treating cancer
CN116253798B (en) * 2022-12-15 2023-10-27 华中农业大学 Neutralizing monoclonal antibody for S1 protein conformational epitope of porcine delta coronavirus
CN116444673B (en) * 2023-06-14 2023-09-15 北京汇智和源生物技术有限公司 Humanized membrane CD14 monoclonal antibody and preparation method and application thereof
CN116735873B (en) * 2023-08-09 2023-10-31 北京纳百生物科技有限公司 Application of monoclonal antibody specifically binding to canine parvovirus VP2 protein in detection reagent

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS58201994A (en) 1982-05-21 1983-11-25 Hideaki Hagiwara Method for producing antigen-specific human immunoglobulin
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
ES2139598T3 (en) 1990-07-10 2000-02-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF SPECIFIC UNION COUPLE MEMBERS.
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
WO1994025585A1 (en) 1993-04-26 1994-11-10 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ES2108048T3 (en) 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
DE69229477T2 (en) 1991-09-23 1999-12-09 Cambridge Antibody Tech Methods for the production of humanized antibodies
AU3178993A (en) 1991-11-25 1993-06-28 Enzon, Inc. Multivalent antigen-binding proteins
WO1993011236A1 (en) 1991-12-02 1993-06-10 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
EP0746609A4 (en) 1991-12-17 1997-12-17 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
JP3507073B2 (en) 1992-03-24 2004-03-15 ケンブリッジ アンティボディー テクノロジー リミティド Methods for producing members of a specific binding pair
ATE381614T1 (en) 1992-07-24 2008-01-15 Amgen Fremont Inc FORMATION OF XENOGENE ANTIBODIES
US5648267A (en) 1992-11-13 1997-07-15 Idec Pharmaceuticals Corporation Impaired dominant selectable marker sequence and intronic insertion strategies for enhancement of expression of gene product and expression vector systems comprising same
GB9313509D0 (en) 1993-06-30 1993-08-11 Medical Res Council Chemisynthetic libraries
AU690171B2 (en) 1993-12-03 1998-04-23 Medical Research Council Recombinant binding proteins and peptides
DE69535243T2 (en) 1994-07-13 2007-05-10 Chugai Seiyaku K.K. AGAINST HUMAN INTERLEUKIN-8 DIRECTED, RECONSTITUTED HUMAN ANTIBODY
KR100654645B1 (en) 1995-04-27 2007-04-04 아브게닉스, 인크. Human Antibodies from Immunized Genomous
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
WO2001019858A2 (en) 1999-09-15 2001-03-22 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A family of ubiquitin-like proteins binds the atpase domain of hsp70-like stch
US7420030B2 (en) 2000-09-08 2008-09-02 The Board Of Regents Of The University Of Texas System Aminopeptidase A (APA) targeting peptides for the treatment of cancer
US7452964B2 (en) 2001-09-07 2008-11-18 Board Of Regents, The University Of Texas System Compositions and methods of use of targeting peptides against placenta and adipose tissues
JP2004515751A (en) * 2000-09-08 2004-05-27 ボード・オブ・リージエンツ,ザ・ユニバーシテイ・オブ・テキサス・システム Methods and compositions for in vitro targeting
US7671010B2 (en) 2002-08-30 2010-03-02 The Board Of Regents Of The University Of Texas System Compositions and methods of use of targeting peptides for diagnosis and therapy of human cancer
WO2003104453A1 (en) 2002-06-05 2003-12-18 中外製薬株式会社 Method of constructing antibody
EP1546714A4 (en) * 2002-08-30 2007-12-05 Univ Texas Compositions and methods of use of targeting peptides for diagnosis and therapy of human cancer
US20050053993A1 (en) 2002-12-18 2005-03-10 Davidson Donald J. Uses of an endothelial cell receptor
WO2005045428A2 (en) * 2003-10-27 2005-05-19 University Of Southern California Methods and compositions for modulating apoptosis
GB0404936D0 (en) * 2004-03-04 2004-04-07 Univ London Screen

Also Published As

Publication number Publication date
CA2679266A1 (en) 2008-09-04
EP2130552A4 (en) 2011-03-02
US8192740B2 (en) 2012-06-05
US20100041074A1 (en) 2010-02-18
EP2130552B1 (en) 2016-06-15
CN101951953A (en) 2011-01-19
JPWO2008105560A1 (en) 2010-06-03
JP5374360B2 (en) 2013-12-25
EP2130552A1 (en) 2009-12-09
WO2008105560A1 (en) 2008-09-04

Similar Documents

Publication Publication Date Title
US8192740B2 (en) Pharmaceutical composition comprising anti-GRP78 antibody as active ingredient
US20210380715A1 (en) Anti-dll3 antibody
US8975374B2 (en) Pharmaceutical composition comprising anti-HB-EGF antibody as active ingredient
CA2669731C (en) Anti-human dlk-1 antibody showing anti-tumor activity in vivo
JP7070932B2 (en) BAFF-R Targeted Chimeric Antigen Receptor Modified T Cells and Their Use
US20100061933A1 (en) Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
AU2009203464A1 (en) Anti-CLDN6 antibody
US20120282173A1 (en) Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
WO2011105573A1 (en) Anti-icam3 antibody and use thereof
CN113474372A (en) Monoclonal antibody of anti-CEACAM 5, preparation method and application thereof
US20120128678A1 (en) Diagnosis and treatment of cancer using anti-tmprss11e antibody
JP2005536199A (en) Antibodies and their use
WO2018235855A1 (en) Antisulfataed glycosaminoglycan antibody
US9416189B2 (en) Anti-CXADR antibody
JP2023511189A (en) SEMG2 Antibodies and Uses Thereof
WO2011052753A1 (en) Antibody binding to mansc1 protein and having anticancer activity
AU2005221470B2 (en) Inhibitors of extracellular hsp90
CN117843793A (en) Anti-mesothelin antibodies, antigen binding fragments and uses thereof
NZ755670B2 (en) Multispecific antibodies, multispecific activatable antibodies and methods of using the same
JP2014162754A (en) Monoclonal antibody against cell membrane protein expressing specifically to cancer stem cell

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application